1
|
Tang Z, Ye J, Chen D. HHLA3 Silencing Suppresses KRAS-Mutant Non-Small-Cell Lung Cancer Cell Progression Through Triggering MYEOV-Mediated Ferroptosis. J Biochem Mol Toxicol 2025; 39:e70271. [PMID: 40262052 DOI: 10.1002/jbt.70271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/11/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
KRAS mutation is one of the most common mutational events in non-small-cell lung cancer (NSCLC). However, due to the complex signaling pathways and high biological heterogeneity of KRAS-mutant NSCLC, the current clinical treatment for patients with KRAS mutations still faces many difficulties. The oncogenic effector in KRAS-mutant NSCLC was screened using GEO data sets. CCK-8, colony formation, transwell, and flow cytometry were conducted to assess the malignant phenotype of KRAS-mutant NSCLC cells. The indicators intracellular Fe2+, ROS, GSH, and MDA levels were employed to reflect the ferroptosis of cells. The mechanism of myeloma overexpressed (MYEOV) in KRAS-mutant NSCLC was explored from the perspective of noncoding RNA (ncRNA) and validated by rescue experiments. MYEOV presented a high expression trend in KRAS-mutant NSCLC specimens. MYEOV silencing effectively repressed the malignant phenotype and promoted ferroptosis of NSCLC cells carrying KRAS mutations. Based on bioinformation analysis and a series of rescue experiments, we established the HHLA3/miR-139-5p/MYEOV regulatory network in KRAS-mutant NSCLC cells and disclosed that HHLA3 served as a molecular sponge for miR-139-5p to regulate MYEOV expression. The mechanism of MYEOV and its ncRNA network affecting the progression of KRAS-mutant NSCLC revealed in this study intends to provide a theoretical basis for KRAS-mutant NSCLC treatment.
Collapse
Affiliation(s)
- Zhimiao Tang
- Department of Cardiothoracic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine (Jinhua Central Hospital), Jinhua, Zhejiang, China
| | - Jia Ye
- Department of Cardiothoracic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine (Jinhua Central Hospital), Jinhua, Zhejiang, China
| | - Dong Chen
- Department of Cardiothoracic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine (Jinhua Central Hospital), Jinhua, Zhejiang, China
| |
Collapse
|
2
|
Liguori L, Salomone F, Viggiano A, Sabbatino F, Pepe S, Formisano L, Bianco R, Servetto A. KRAS mutations in advanced non-small cell lung cancer: From biology to novel therapeutic strategies. Crit Rev Oncol Hematol 2025; 205:104554. [PMID: 39522850 DOI: 10.1016/j.critrevonc.2024.104554] [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: 05/16/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Kristen rat sarcoma viral oncogene homolog (KRAS) mutations play a major role in the carcinogenesis of many types of solid tumors including non-small cell lung cancer (NSCLC). Among KRAS mutations, p.G12C single-nucleotide variant (KRASG12C) is the most frequently reported in NSCLC patients, with a prevalence of about 12-13 %. For many decades, KRAS mutations including KRASG12C were considered "undruggable" because of the lack of effective and well-tolerated selective therapies. Noteworthy, CodeBreaK100 and KRYSTAL-1 clinical trials have recently demonstrated that sotorasib and adagrasib, two novel selective KRASG12C inhibitors, have clinical activity with acceptable adverse-event profile for the treatment of advanced NSCLC patients with KRASG12C mutation. On the other hand, no selective therapies are approved for the treatment of advanced NSCLC patients with non-G12C KRAS mutations. As a result, these patients receive the same treatments as those without KRAS mutations. In this paper, we describe the role of KRAS mutations in NSCLC focusing on the clinical and molecular characteristics which potentially identify specific subtypes of NSCLC patients based on different KRAS mutations. We also provide an overview of the main clinical trials testing novel selective KRASG12C inhibitors as well as novel potential therapeutic strategies for NSCLC patients with non-G12C KRAS mutations.
Collapse
Affiliation(s)
- Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy; Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84031, Italy.
| | - Fabio Salomone
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| | - Angela Viggiano
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy
| | - Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84031, Italy.
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84031, Italy.
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| |
Collapse
|
3
|
Wang R, Liao Z, Liu C, Yu S, Xiang K, Wu T, Feng J, Ding S, Yu T, Cheng G, Li S. CRABP2 promotes cell migration and invasion by activating PI3K/AKT and MAPK signalling pathways via upregulating LAMB3 in prostate cancer. J Biochem 2024; 176:313-324. [PMID: 39038078 DOI: 10.1093/jb/mvae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
Prostate cancer (PCa) has become a worldwide health burden among men. Previous studies have suggested that cellular retinoic acid binding protein 2 (CRABP2) significantly affects the regulation of cell proliferation, motility and apoptosis in multiple cancers; however, the effect of CRABP2 on PCa is poorly reported. CRABP2 expression in different PCa cell lines and its effect on different cellular functions varied. While CRABP2 promotes cell migration and invasion, it appears to inhibit cell proliferation specifically in PC-3 cells. However, the proliferation of DU145 and 22RV1 cells did not appear to be significantly affected by CRABP2. Additionally, CRABP2 had no influence on the cell cycle distribution of PCa cells. The RNA-seq assay showed that overexpressing CRABP2 upregulated laminin subunit beta-3 (LAMB3) mRNA expression, and the enrichment analyses revealed that the differentially expressed genes were enriched in the phosphoinositide 3-kinase (PI3K)/activated protein kinase B (AKT) and mitogen-activated protein kinase (MAPK) signalling pathways. The following western blot experiments also confirmed the upregulated LAMB3 protein level and the activation of the PI3K/AKT and MAPK signalling pathways. Moreover, overexpressing CRABP2 significantly inhibited tumour growth in vivo. In conclusion, CRABP2 facilitates cell migration and invasion by activating PI3K/AKT and MAPK signalling pathways through upregulating LAMB3 in PCa.
Collapse
Affiliation(s)
- Rui Wang
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Zhaoping Liao
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Chunhua Liu
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Shifang Yu
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Kaihua Xiang
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Ting Wu
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Jie Feng
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Senjuan Ding
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Tingao Yu
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Gang Cheng
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| | - Sanlian Li
- Department of Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
4
|
Yu F, Zeng G, Yang L, Zhou H, Wang Y. LAMB3: Central role and clinical significance in neoplastic and non-neoplastic diseases. Biomed Pharmacother 2024; 178:117233. [PMID: 39111076 DOI: 10.1016/j.biopha.2024.117233] [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: 05/25/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
Recently, topics related to targeted gene therapy and diagnosis have become increasingly important in disease research. The progression of many diseases is associated with specific gene signaling pathways. Therefore, the identification of precise gene targets in various diseases is crucial for the development of effective treatments. Laminin subunit beta 3 (LAMB3), a component of laminin 5, functions as an adhesive protein in the extracellular matrix and plays a vital role in regulating cell proliferation, migration, and cell cycle in certain diseases. Previous studies have indicated that LAMB3 is highly expressed in numerous tumorous and non-tumorous conditions, including renal fibrosis; squamous cell carcinoma of the skin, thyroid, lung, pancreatic, ovarian, colorectalr, gastric, breast, cervical, nasopharyngeal, bladder, prostate cancers; and cholangiocarcinoma. Conversely, it is underexpressed in other conditions, such as hepatocellular carcinoma, epidermolysis bullosa, and amelogenesis imperfecta. Consequently, LAMB3 may serve as a molecular diagnostic and therapeutic target for various diseases through its involvement in critical gene signaling pathways. This paper reviews the research status of LAMB3 and its role in related diseases.
Collapse
Affiliation(s)
- Fangqiu Yu
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Guoqiang Zeng
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Lei Yang
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Honglan Zhou
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Yuantao Wang
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China.
| |
Collapse
|
5
|
Zhu C, Guan X, Zhang X, Luan X, Song Z, Cheng X, Zhang W, Qin JJ. Targeting KRAS mutant cancers: from druggable therapy to drug resistance. Mol Cancer 2022; 21:159. [PMID: 35922812 PMCID: PMC9351107 DOI: 10.1186/s12943-022-01629-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) is the most frequently mutated oncogene, occurring in a variety of tumor types. Targeting KRAS mutations with drugs is challenging because KRAS is considered undruggable due to the lack of classic drug binding sites. Over the past 40 years, great efforts have been made to explore routes for indirect targeting of KRAS mutant cancers, including KRAS expression, processing, upstream regulators, or downstream effectors. With the advent of KRAS (G12C) inhibitors, KRAS mutations are now druggable. Despite such inhibitors showing remarkable clinical responses, resistance to monotherapy of KRAS inhibitors is eventually developed. Significant progress has been made in understanding the mechanisms of drug resistance to KRAS-mutant inhibitors. Here we review the most recent advances in therapeutic approaches and resistance mechanisms targeting KRAS mutations and discuss opportunities for combination therapy.
Collapse
Affiliation(s)
- Chunxiao Zhu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.,School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.,Key Laboratory of Prevention, Diagnosis, and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Xinuo Zhang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.,College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhengbo Song
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Xiangdong Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China. .,Key Laboratory of Prevention, Diagnosis, and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Jiang-Jiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China. .,School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China. .,Key Laboratory of Prevention, Diagnosis, and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
| |
Collapse
|
6
|
FAK in Cancer: From Mechanisms to Therapeutic Strategies. Int J Mol Sci 2022; 23:ijms23031726. [PMID: 35163650 PMCID: PMC8836199 DOI: 10.3390/ijms23031726] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 01/25/2023] Open
Abstract
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is overexpressed and activated in many cancer types. FAK regulates diverse cellular processes, including growth factor signaling, cell cycle progression, cell survival, cell motility, angiogenesis, and the establishment of immunosuppressive tumor microenvironments through kinase-dependent and kinase-independent scaffolding functions in the cytoplasm and nucleus. Mounting evidence has indicated that targeting FAK, either alone or in combination with other agents, may represent a promising therapeutic strategy for various cancers. In this review, we summarize the mechanisms underlying FAK-mediated signaling networks during tumor development. We also summarize the recent progress of FAK-targeted small-molecule compounds for anticancer activity from preclinical and clinical evidence.
Collapse
|
7
|
Rahman S, Garrel S, Gerber M, Maitra R, Goel S. Therapeutic Targets of KRAS in Colorectal Cancer. Cancers (Basel) 2021; 13:6233. [PMID: 34944853 PMCID: PMC8699097 DOI: 10.3390/cancers13246233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/28/2022] Open
Abstract
Patients with metastatic colorectal cancer have a 5-year overall survival of less than 10%. Approximately 45% of patients with metastatic colorectal cancer harbor KRAS mutations. These mutations not only carry a predictive role for the absence of response to anti-EGFR therapy, but also have a negative prognostic impact on the overall survival. There is a growing unmet need for a personalized therapy approach for patients with KRAS-mutant colorectal cancer. In this article, we focus on the therapeutic strategies targeting KRAS- mutant CRC, while reviewing and elaborating on the discovery and physiology of KRAS.
Collapse
Affiliation(s)
- Shafia Rahman
- Department of Medical Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, 1695 Eastchester Road Bronx, New York, NY 10461, USA; (S.R.); (R.M.)
| | - Shimon Garrel
- Department of Biology, Lander College For Men, 75-31 150th Street, Flushing, New York, NY 11367, USA;
| | - Michael Gerber
- Department of Biology, Yeshiva University, 500 West 185th Street, New York, NY 10033, USA;
| | - Radhashree Maitra
- Department of Medical Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, 1695 Eastchester Road Bronx, New York, NY 10461, USA; (S.R.); (R.M.)
- Department of Biology, Yeshiva University, 500 West 185th Street, New York, NY 10033, USA;
| | - Sanjay Goel
- Department of Medical Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, 1695 Eastchester Road Bronx, New York, NY 10461, USA; (S.R.); (R.M.)
| |
Collapse
|
8
|
Liu J, Xue L, Xu X, Luo J, Zhang S. FAK-targeting PROTAC demonstrates enhanced antitumor activity against KRAS mutant non-small cell lung cancer. Exp Cell Res 2021; 408:112868. [PMID: 34648846 DOI: 10.1016/j.yexcr.2021.112868] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 01/30/2023]
Abstract
Focal adhesion kinase (FAK) has been established as a promising therapeutic target for KRAS mutant non-small cell lung cancer (NSCLC). However, phase II clinical trials of a FAK inhibitor (Defactinib) have only shown modest antitumor activity. To address this challenge, here we report the use of a FAK-targeting proteolysis targeting chimera (D-PROTAC) to treat KRAS mutant NSCLC. We validated that D-PROTAC could efficiently eliminate FAK protein via the ubiquitin-proteasome pathway in KRAS mutant NSCLC A427 cells, causing over 90% degradation at 800 nM. After comparing both in vitro and in vivo therapeutic efficacies, we demonstrated that D-PRTOAC outperformed Defactinib in inhibiting tumor growth. Specifically, D-PROTAC at 800 nM reduced cell viability, migration, and invasion by ∼80%. Furthermore, a ∼85% suppression of tumor growth was elicited by D-PROTAC when intratumorally administrated at 10 mg/kg in subcutaneous A427-bearing mice. These results thus demonstrate for the first time that PROTACs may serve as promising therapeutic agents for the intractable NSCLC harboring KRAS mutations.
Collapse
Affiliation(s)
- Jinyuan Liu
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lei Xue
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiang Xu
- Taixing People's Hospital, Taixing, 225400, China
| | - Jinhua Luo
- Department of Thoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Shijiang Zhang
- Department of Cardiothoracic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
9
|
Ma J, Xu LY, Sun QH, Wan XY, BingLi. Inhibition of miR-1298-5p attenuates sepsis lung injury by targeting SOCS6. Mol Cell Biochem 2021; 476:3745-3756. [PMID: 34100174 DOI: 10.1007/s11010-021-04170-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Sepsis is one of the leading causes of morbidity and mortality and a major cause of acute lung injury (ALI). carried by exosomes play a role in a variety of diseases. However,there are not many studies of exosomal miRNAs in sepsis and sepsis lung injury.miR-1298-5p and suppressor of cytokine signaling 6 (SOCS6) were silenced or overexpressed in human bronchial epithelial cells (BEAS-2B). PKH-67 Dye was used to trace exosome endocytosis. Cell permeability was evaluated by measuring trans-epithelial electrical resistance (TEER) and FITC dextran flux. ELISA kits were used for cytokine detection. Quantitative RT-PCR and western blots were used to evaluate gene expression. miR-1298-5p was elevated in exosomes from patients with sepsis lung injury (Sepsis_exo). Treatment of BEAS-2B cells using Sepsis_exo significantly inhibited cell proliferation, and induced cell permeability and inflammatory response. miR-1298-5p directly targeted SOCS6. Overexpressing SOCS6 reversed miR-1298-5p-induced cell permeability and inflammatory response. Inhibition of STAT3 blocked SOCS6-silencing caused significant increase of cell permeability and inflammation. Exosomes isolated from patients of sepsis lung injury increased cell permeability and inflammatory response in BEAS-2B cells through exosomal miR-1298-5p which targeted SOCS6 via STAT3 pathway. The findings highlight the importance of miR-1298-5p/SOCS6/STAT3 axis in sepsis lung injury and provide new insights into therapeutic strategies for sepsis lung injury.
Collapse
Affiliation(s)
- Jian Ma
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Doctor's Office, 10th floor, building 2, NO.507 Zhengmin Road, Yangpu District, Shanghai, 200433, P.R. China.
| | - Li-Yun Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Doctor's Office, 10th floor, building 2, NO.507 Zhengmin Road, Yangpu District, Shanghai, 200433, P.R. China
| | - Qiu-Hong Sun
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Doctor's Office, 10th floor, building 2, NO.507 Zhengmin Road, Yangpu District, Shanghai, 200433, P.R. China
| | - Xiao-Yu Wan
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Doctor's Office, 10th floor, building 2, NO.507 Zhengmin Road, Yangpu District, Shanghai, 200433, P.R. China
| | - BingLi
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Doctor's Office, 10th floor, building 2, NO.507 Zhengmin Road, Yangpu District, Shanghai, 200433, P.R. China
| |
Collapse
|
10
|
Chen P, Qin Z, Sun X, Yang J, Lv J, Diao M. Expression and clinical significance of lncRNA OSER1-AS1 in peripheral blood of patients with non-small cell lung cancer. Cells Tissues Organs 2021; 211:589-600. [PMID: 34525476 DOI: 10.1159/000519529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/05/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Peirui Chen
- Department of Cardiothoracic Surgery, People's Hospital of Deyang City, Deyang City, China
| | - Zheng Qin
- Department of Cardiothoracic Surgery, People's Hospital of Deyang City, Deyang City, China
| | - Xiaokang Sun
- Department of Cardiothoracic Surgery, People's Hospital of Deyang City, Deyang City, China
| | - Junrong Yang
- Department of Cardiothoracic Surgery, People's Hospital of Deyang City, Deyang City, China
| | - Jing Lv
- Department of Cardiothoracic Surgery, People's Hospital of Deyang City, Deyang City, China
| | - Mingqiang Diao
- Department of Cardiothoracic Surgery, People's Hospital of Deyang City, Deyang City, China
| |
Collapse
|
11
|
Crosstalk between Environmental Inflammatory Stimuli and Non-Coding RNA in Cancer Occurrence and Development. Cancers (Basel) 2021; 13:cancers13174436. [PMID: 34503246 PMCID: PMC8430834 DOI: 10.3390/cancers13174436] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/20/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Increasing evidence has indicated that chronic inflammatory processes have an influence on tumor occurrence and all stages of tumor development. A dramatic increase of studies into non-coding RNAs (ncRNAs) biology has shown that ncRNAs act as oncogenic drivers and tumor suppressors in various inflammation-induced cancers. Thus, this complex network of inflammation-associated cancers and ncRNAs offers targets for prevention from the malignant transformation from inflammation and treatment of malignant diseases. Abstract There is a clear relationship between inflammatory response and different stages of tumor development. Common inflammation-related carcinogens include viruses, bacteria, and environmental mutagens, such as air pollutants, toxic metals, and ultraviolet light. The expression pattern of ncRNA changes in a variety of disease conditions, including inflammation and cancer. Non-coding RNAs (ncRNAs) have a causative role in enhancing inflammatory stimulation and evading immune responses, which are particularly important in persistent pathogen infection and inflammation-to-cancer transformation. In this review, we investigated the mechanism of ncRNA expression imbalance in inflammation-related cancers. A better understanding of the function of inflammation-associated ncRNAs may help to reveal the potential of ncRNAs as a new therapeutic strategy.
Collapse
|
12
|
Zhang B, Zhang Y, Zhang J, Liu P, Jiao B, Wang Z, Ren R. Focal Adhesion Kinase (FAK) Inhibition Synergizes with KRAS G12C Inhibitors in Treating Cancer through the Regulation of the FAK-YAP Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100250. [PMID: 34151545 PMCID: PMC8373085 DOI: 10.1002/advs.202100250] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/20/2021] [Indexed: 05/04/2023]
Abstract
KRAS mutation is one of the most prevalent genetic drivers of cancer development, yet KRAS mutations are until very recently considered undruggable. There are ongoing trials of drugs that target the KRAS G12C mutation, yet acquired drug resistance from the extended use has already become a major concern. Here, it is demonstrated that KRAS G12C inhibition induces sustained activation of focal adhesive kinase (FAK) and show that a combination therapy comprising KRAS G12C inhibition and a FAK inhibitor (IN10018) achieves synergistic anticancer effects. It can simultaneously reduce the extent of drug resistance. Diverse CDX and PDX models of KRAS G12C mutant cancer are examined and synergistic benefits from the combination therapy are consistently observed. Mechanistically, it is found that both aberrant FAK-YAP signaling and FAK-related fibrogenesis impact on the development of KRAS G12C inhibitor resistance. This study thus illustrates the mechanism of resistance of cancer to the treatment of KRAS G12C inhibitor, as well as an innovative combination therapy to improve treatment outcomes for KRAS G12C mutant cancers.
Collapse
Affiliation(s)
- Baoyuan Zhang
- Shanghai Institute of HematologyState Key Laboratory for Medical GenomicsNational Research Center for Translational MedicineInternational Center for Aging and CancerCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yan Zhang
- Shanghai Institute of HematologyState Key Laboratory for Medical GenomicsNational Research Center for Translational MedicineInternational Center for Aging and CancerCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | | | - Ping Liu
- Shanghai Institute of HematologyState Key Laboratory for Medical GenomicsNational Research Center for Translational MedicineInternational Center for Aging and CancerCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Bo Jiao
- Shanghai Institute of HematologyState Key Laboratory for Medical GenomicsNational Research Center for Translational MedicineInternational Center for Aging and CancerCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zaiqi Wang
- InxMed (Shanghai) Co., LtdShanghai201202China
| | - Ruibao Ren
- Shanghai Institute of HematologyState Key Laboratory for Medical GenomicsNational Research Center for Translational MedicineInternational Center for Aging and CancerCollaborative Innovation Center of HematologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
13
|
Guan H, Sun C, Gu Y, Li J, Ji J, Zhu Y. Circular RNA circ_0003028 contributes to tumorigenesis by regulating GOT2 via miR-1298-5p in non-small cell lung cancer. Bioengineered 2021; 12:2326-2340. [PMID: 34077306 PMCID: PMC8806680 DOI: 10.1080/21655979.2021.1935064] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common malignant tumor, with high morbidity and mortality. Circular RNA (circRNA) circ_0003028 was reported to be upregulated in NSCLC. This study is designed to explore the role and mechanism of circ_0003028 on NSCLC progression. In this work, circ_0003028, microRNA-1298-5p (miR-1298-5p), and glutamic oxaloacetic transaminase 2 (GOT2) level were detected by real-time quantitative polymerase chain reaction (RT-qPCR). The localization of circ_0003028 was analyzed by subcellular fractionation assay. Cell proliferation, colony number, cell cycle progression, apoptosis, migration, invasion, and angiogenesis were measured by Cell Counting Kit-8 (CCK-8), colony formation, flow cytometry, transwell, and tube formation assays. Protein levels of Beclin1, light chain 3 (LC3)-II/LC3-I, GOT2, proliferating cell nuclear antigen (PCNA) were examined by western blot assay. The binding relationship between miR-1298-5p and circ_0003028 or GOT2 was predicted by circular RNA Interactome or starbase and then verified by dual-luciferase reporter, RNA Immunoprecipitation (RIP), and RNA pull-down assays. The biological role of circ_0003028 on NSCLC tumor growth was examined by the xenograft tumor model in vivo. We reported that circ_0003028 and GOT2 were upregulated, and miR-1298-5p was decreased in NSCLC tissues and cells. Moreover, circ_0003028 knockdown curbed cell proliferative ability, migration, invasion, angiogenesis, and facilitate apoptosis and autophagy in NSCLC cells in vitro. Mechanical analysis discovered that circ_0003028 regulated GOT2 expression by sponging miR-1298-5p. Circ_0003028 silencing hindered the cell growth of NSCLC in vivo. Taken together, circ_0003028 knockdown could suppress NSCLC progression partly by regulating the miR-1298-5p/GOT2 axis, providing an underlying therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Hongjun Guan
- Department of Thoracic Surgery, Jianhu Hospital Affiliated to Nantong University, Yancheng, China
| | - Changpeng Sun
- Department of Thoracic Surgery, Jianhu Hospital Affiliated to Nantong University, Yancheng, China
| | - Yinfeng Gu
- Department of Thoracic Surgery, Jianhu Hospital Affiliated to Nantong University, Yancheng, China
| | - Jinjin Li
- Department of Thoracic Surgery, Jianhu Hospital Affiliated to Nantong University, Yancheng, China
| | - Jie Ji
- Information Center, Jianhu Hospital Affiliated to Nantong University, Yancheng, China
| | - Yongxian Zhu
- Department of Thoracic Surgery, Jianhu Hospital Affiliated to Nantong University, Yancheng, China
| |
Collapse
|
14
|
Zhang H, Zhang R, Zhang G, Liu W, Ma Z, Yue C, Yang M. Clinical significance of miR-1298 in cervical cancer and its biological function in vitro. Oncol Lett 2021; 21:401. [PMID: 33777224 PMCID: PMC7988695 DOI: 10.3892/ol.2021.12662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
Cervical cancer is one of the most malignant tumors in women. miR-1298 was reported to be abnormally expressed and serve crucial role in tumorigenesis of several types of cancer; however, the role of miR-1298 in cervical cancer remains unknown. The present study aimed to evaluate the clinical and biological significance of miR-1298 in cervical cancer. To do so, the expression level of miR-1298 in cervical cancer tissues and cells was evaluated by reverse transcription quantitative PCR. Kaplan-Meier survival analysis and Cox regression analysis were used to explore the prognostic significance of miR-1298 in patients with cervical cancer. Cell Counting Kit-8 and Transwell migration and invasion assays were used to evaluate the effect of miR-1298 on the proliferative, migratory and invasive abilities of cervical cancer cells, respectively. The expression of miR-1298 was lower in cancer tissues and cells compared with normal tissues and cells. Furthermore, miR-1298 expression was associated with lymph node metastasis, tumor diameter and staging from the International Federation of Gynecology and Obstetrics. In addition, patients with low miR-1298 expression had poorer overall survival. These findings suggested that miR-1298 may be considered as an independent prognostic factor for patients with cervical cancer. Furthermore, the results demonstrated that miR-1298 knockdown could promote tumor cell proliferation and migratory and invasive abilities. In addition, nucleus accumbens-associated 1 (NACC1) was demonstrated to be a direct target of miR-1298. Taken together, these findings indicated that miR-1298 overexpression may be considered as a prognostic biomarker for cervical cancer and that miR-1298 may play an inhibitor role in cervical cancer by targeting NACC1.
Collapse
Affiliation(s)
- Haitao Zhang
- Department of Pathology, Chengwu People's Hospital, Heze, Shandong 274200, P.R. China
| | - Ruihong Zhang
- Department of Obstetrics, Chengwu People's Hospital, Heze, Shandong 274200, P.R. China
| | - Guiling Zhang
- Department of Pathology, Chengwu People's Hospital, Heze, Shandong 274200, P.R. China
| | - Wenjuan Liu
- Department of Pathology, Chengwu People's Hospital, Heze, Shandong 274200, P.R. China
| | - Zhaoyuan Ma
- Department of Pathology, Linyi Hot Spring Sanatorium of Shandong Coal Industry Bureau, Linyi, Shandong 276032, P.R. China
| | - Caiyun Yue
- Department of Pathology, Chengwu People's Hospital, Heze, Shandong 274200, P.R. China
| | - Min Yang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
15
|
microRNA-1298 inhibits the malignant behaviors of breast cancer cells via targeting ADAM9. Biosci Rep 2021; 40:226894. [PMID: 33146718 PMCID: PMC7729294 DOI: 10.1042/bsr20201215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) regulate the progression of human malignancy by targeting oncogenes or tumor suppressors, which are 12 promising targets for cancer treatment. Increasing evidence has suggested the aberrant expression and tumor-suppressive function of miR-1298 in cancers, however, the regulatory mechanism of miR-1298 in breast cancer (BC) remains unclear. Here, our findings showed that miR-1298 was down-regulated in BC tissues and cell lines. Lower level of miR-1298 was significantly correlated with the advanced progression of BC patients. Experimental study showed that overexpression of miR-1298 inhibited the proliferation, induced apoptosis and cell cycle arrest in BC cells. The in vivo xenograft mice model showed that highly expressed miR-1298 significantly reduced the tumor growth and metastasis. Further mechanism analysis revealed that miR-1298 bound the 3′-untranslated region (UTR) of a disintegrin and metalloproteinase 9 domain (ADAM9) and suppressed the expression of ADAM9 in BC cells. ADAM9 was overexpressed in BC tissues and inversely correlated with miR-1298. Down-regulation of ADAM9 induced apoptosis and cell cycle arrest of BC cells. Moreover, ectopic expression of ADAM9 by transiently transfecting with vector encoding the full coding sequence of ADAM9 attenuated the inhibitory effects of miR-1298 on the proliferation and cell cycle progression of BC cells. Collectively, our results illustrated that miR-1298 played a suppressive role in regulating the phenotype of BC cells through directly repressing ADAM9, suggesting the potential application of miR-1298 in the therapy of BC.
Collapse
|
16
|
Zhang J, Hu D. miR-1298-5p Influences the Malignancy Phenotypes of Breast Cancer Cells by Inhibiting CXCL11. Cancer Manag Res 2021; 13:133-145. [PMID: 33469358 PMCID: PMC7810718 DOI: 10.2147/cmar.s279121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/11/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Breast cancer (BC) has deleterious effects on women's health worldwide, yet its molecular mechanism remains unclear. OBJECTIVE This study aimed to discover the underlying mechanism used by miR-1298-5p to regulate CXCL11 in BC. METHODS Microarray analysis was performed to identify the key mRNA and miRNA involved in BC. The expression of miR-1298-5p and CXCL11 mRNA in BC clinical tissues and cell lines was detected using quantitative reverse transcription PCR (RT-qPCR), while the demonstration of intra- and extra-cellular CXCL11 protein was measured using western-blotting or ELISA assay. CCK-8, BrdU ELISA, colony formation, wound healing, and cell adhesion assays were carried out to determine cell viability, cell proliferation, colony formation, cell migration and adhesion phenotypes, respectively. A dual-luciferase assay kit was also employed to confirm the predicted binding scheme between miR-1298-5p and CXCL11. RESULTS Microarray analysis confirmed miR-1298-5p and CXCL11 as the miRNA and mRNA to be further investigated in BC. After observing low-level miR-1298-5p and high-level CXCL11 in BC clinical tissues and cell lines, it was discovered that miR-1298-5p inhibited the phenotypes of BC cells, while CXCL11 promoted the tumorigenesis of BC cells. Findings indicated that miR-1298-5p attenuated the promotive effect of CXCL11 on BC cell phenotypes. CONCLUSION This research revealed that miR-1298-5p could influence the malignancy phenotypes of BC cells by inhibiting CXCL11.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Breast Surgery, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei067000, People’s Republic of China
| | - Dawei Hu
- Department of Breast Surgery, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei067000, People’s Republic of China
| |
Collapse
|
17
|
Liu X, Ju J, Liu Q, Zhu Z, Liu C. The Chinese Medicine, Shezhi Huangling Decoction, Inhibits the Growth and Metastasis of Glioma Cells via the Regulation of miR-1298-5p/TGIF1 Axis. Cancer Manag Res 2020; 12:5677-5687. [PMID: 32765071 PMCID: PMC7367721 DOI: 10.2147/cmar.s242856] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose In recent years, traditional Chinese medicine has achieved good results in treating gliomas. This research aimed to reveal the effect of Shezhi Huangling decoction (SD) on glioma cell process. Methods U87 and U251 cells were treated with different concentrations (10, 30 and 50 μg/mL) of SD or transfected with miR-1298-5p mimic, inhibitor and siRNA targeting TGIF1. Cell proliferation, migration, invasion and apoptosis were detected. The expression of miR-1298-5p was measured by qRT-PCR, while TGIF1 expression was examined by immunohistochemical analysis and Western blot. Results SD treatment inhibited the proliferation, migration and invasion of glioma cells and induced the apoptosis. In addition, SD treatment induced the expression of miR-1298-5p in glioma cells. The low expression of miR-1298-5p was examined in glioma tissues and was significantly related to the high histological grade of glioma patients and predicted a poor prognosis. MiR-1298-5p directly targeted the 3'-UTR of transforming growth factor β induced factor 1 (TGIF1) and reduced TGIF1 protein expression. MiR-1298-5p restricted the proliferation, migration and invasion of glioma cells and induced cell apoptosis by targeting TGIF1. Conclusion Our data reveal that SD acts as a cancer-inhibiting agent in glioma via miR-1298-5p/TGIF1 axis, suggesting a potential therapeutic application of SD in glioma.
Collapse
Affiliation(s)
- Xiaoqian Liu
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Jianfeng Ju
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Qun Liu
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Zongmin Zhu
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Chunxia Liu
- Department of Pharmacy, Caoxian People's Hospital of Heze City, Heze 274400, Shandong, People's Republic of China
| |
Collapse
|
18
|
Regulation of DNA Damage Response and Homologous Recombination Repair by microRNA in Human Cells Exposed to Ionizing Radiation. Cancers (Basel) 2020; 12:cancers12071838. [PMID: 32650508 PMCID: PMC7408912 DOI: 10.3390/cancers12071838] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Ionizing radiation may be of both artificial and natural origin and causes cellular damage in living organisms. Radioactive isotopes have been used significantly in cancer therapy for many years. The formation of DNA double-strand breaks (DSBs) is the most dangerous effect of ionizing radiation on the cellular level. After irradiation, cells activate a DNA damage response, the molecular path that determines the fate of the cell. As an important element of this, homologous recombination repair is a crucial pathway for the error-free repair of DNA lesions. All components of DNA damage response are regulated by specific microRNAs. MicroRNAs are single-stranded short noncoding RNAs of 20–25 nt in length. They are directly involved in the regulation of gene expression by repressing translation or by cleaving target mRNA. In the present review, we analyze the biological mechanisms by which miRNAs regulate cell response to ionizing radiation-induced double-stranded breaks with an emphasis on DNA repair by homologous recombination, and its main component, the RAD51 recombinase. On the other hand, we discuss the ability of DNA damage response proteins to launch particular miRNA expression and modulate the course of this process. A full understanding of cell response processes to radiation-induced DNA damage will allow us to develop new and more effective methods of ionizing radiation therapy for cancers, and may help to develop methods for preventing the harmful effects of ionizing radiation on healthy organisms.
Collapse
|
19
|
Zhu Z, Song J, Guo Y, Huang Z, Chen X, Dang X, Huang Y, Wang Y, Ou W, Yang Y, Yu W, Liu CY, Cui L. LAMB3 promotes tumour progression through the AKT-FOXO3/4 axis and is transcriptionally regulated by the BRD2/acetylated ELK4 complex in colorectal cancer. Oncogene 2020; 39:4666-4680. [PMID: 32398865 DOI: 10.1038/s41388-020-1321-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022]
Abstract
Aberrant expression of laminin-332 promotes tumour growth and metastasis in multiple cancers. However, the dysregulated expression and mechanism of action of LAMB3, which encodes the β3 subunit of laminin-332, and the mechanism underlying dysregulated LAMB3 expression in CRC remain obscure. Here, we show that LAMB3 is overexpressed in CRC and that this overexpression is correlated with tumour metastasis and poor prognosis. Overexpression of LAMB3 promoted cell proliferation and cell migration in vitro and tumour growth and metastasis in vivo, while knockdown of LAMB3 elicited opposing effects. LAMB3 inhibited the tumour suppressive function of FOXO3/4 by activating AKT in CRC. Both the BET inhibitor JQ1 and the MEK inhibitor U0126 decreased the mRNA level of LAMB3 in multiple CRC cells. Mechanistically, ELK4 cooperated with BRD2 to regulate the transcription of LAMB3 in CRC by directly binding to the ETS binding motifs in the LAMB3 promoter. ELK4 was as acetylated at K125, which enhanced the interaction between ELK4 and BRD2. JQ1 disrupted the interaction between ELK4 and BRD2, resulting in decreased binding of BRD2 to the LAMB3 promoter and downregulation of LAMB3 transcription. Both ELK4 and BRD2 expression was associated with LAMB3 expression in CRC. LAMB3 expression was also negatively correlated with FOXO3/4 in CRC. Our study reveals the pro-tumorigenic role of LAMB3 through the AKT-FOXO3/4 axis and the transcriptional mechanism of LAMB3 in CRC, demonstrating that LAMB3 is a potential therapeutic target that can be targeted by BET inhibitors and MEK inhibitors.
Collapse
Affiliation(s)
- Zhehui Zhu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China
| | - Jinglue Song
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Yuegui Guo
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Zhenyu Huang
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China
| | - Xiaojian Chen
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China
| | - Xuening Dang
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China
| | - Yuji Huang
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China
| | - Yuhan Wang
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Weijun Ou
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Center for Systems Medicine Research, Chinese Academy of Medical Sciences, 215123, Suzhou, Jiangsu, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Zhongshan Hospital, Fudan University, 200438, Shanghai, China.
| | - Chen-Ying Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China.
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China.
| | - Long Cui
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China.
- Shanghai Colorectal Cancer Research Center, 200092, Shanghai, China.
| |
Collapse
|
20
|
Rousselle P, Scoazec JY. Laminin 332 in cancer: When the extracellular matrix turns signals from cell anchorage to cell movement. Semin Cancer Biol 2020; 62:149-165. [PMID: 31639412 DOI: 10.1016/j.semcancer.2019.09.026] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/22/2019] [Accepted: 09/29/2019] [Indexed: 02/07/2023]
Abstract
Laminin 332 is crucial in the biology of epithelia. This large extracellular matrix protein consists of the heterotrimeric assembly of three subunits - α3, β3, and γ2 - and its multifunctionality relies on a number of extracellular proteolytic processing events. Laminin 332 is central to normal epithelium homeostasis by sustaining cell adhesion, polarity, proliferation, and differentiation. It also supports a major function in epithelial tissue formation, repair, and regeneration by buttressing cell migration and survival and basement membrane assembly. Interest in this protein increased after the discovery that its expression is perturbed in tumor cells, cancer-associated fibroblasts, and the tumor microenvironment. This review summarizes current knowledge regarding the established involvement of the laminin 332 γ2 chain in tumor invasiveness and discusses the role of its α3 and β3 subunits.
Collapse
Affiliation(s)
- Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS - Université Lyon 1, Institut de Biologie et Chimie des Protéines, SFR BioSciences Gerland-Lyon Sud, 7 passage du Vercors, F-69367, France.
| | - Jean Yves Scoazec
- Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif cedex, France; Université Paris Sud, Faculté de Médecine de Bicêtre, 94270 Le Kremlin Bicêtre, France
| |
Collapse
|
21
|
Martini G, Ciardiello D, Vitiello PP, Napolitano S, Cardone C, Cuomo A, Troiani T, Ciardiello F, Martinelli E. Resistance to anti-epidermal growth factor receptor in metastatic colorectal cancer: What does still need to be addressed? Cancer Treat Rev 2020; 86:102023. [PMID: 32474402 DOI: 10.1016/j.ctrv.2020.102023] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022]
Abstract
Colorectal cancer (CRC) represents a global health problem, being one of the most diagnosed and aggressive tumors. Cetuximab and panitumumab monoclonal antibodies (mAbs) in combination with chemotherapy are an effective strategy for patients with RAS Wild Type (WT) metastatic colorectal cancer (mCRC). However, tumors are often unresponsive or develop resistance. In the last years, molecular alterations in principal oncogenes (RAS, BRAF, PI3KCA, PTEN) in the downstream pathway of the epidermal growth factor receptor (EGFR) and in other receptors (HER2, MET) that converge on MAPK-ERK signalling have been identified as novel mechanisms of resistance to anti-EGFR strategies. However, further efforts are needed to better stratify CRCs and ensure more individualized treatments. Herein, we describe the consolidated molecular drivers of resistance and the therapeutic strategies available so far, with an overview on potential biomarkers of response that could be integrated in clinical practice.
Collapse
Affiliation(s)
- Giulia Martini
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Pietro Paolo Vitiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Claudia Cardone
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Antonio Cuomo
- Gastroenterology Unit, Ospedale Umberto I, Nocera Inferiore, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
22
|
Takagawa Y, Gen Y, Muramatsu T, Tanimoto K, Inoue J, Harada H, Inazawa J. miR-1293, a Candidate for miRNA-Based Cancer Therapeutics, Simultaneously Targets BRD4 and the DNA Repair Pathway. Mol Ther 2020; 28:1494-1505. [PMID: 32320642 DOI: 10.1016/j.ymthe.2020.04.001] [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] [Received: 11/20/2019] [Revised: 03/04/2020] [Accepted: 04/02/2020] [Indexed: 12/18/2022] Open
Abstract
BRD4, a member of the bromodomain and extra-terminal domain (BET) protein family, plays a role in the organization of super-enhancers and transcriptional activation of oncogenes in cancer and is recognized as a promising target for cancer therapy. microRNAs (miRNAs), endogenous small noncoding RNAs, cause mRNA degradation or inhibit protein translation of their target genes by binding to complementary sequences. miRNA mimics simultaneously targeting several tumor-promoting genes and BRD4 may be useful as therapeutic agents of tumor-suppressive miRNAs (TS-miRs) for cancer therapy. To investigate TS-miRs for the development of miRNA-based cancer therapeutics, we performed function-based screening in 10 cancer cell lines with a library containing 2,565 human miRNA mimics. Consequently, miR-1293, miR-876-3p, and miR-6571-5p were identified as TS-miRs targeting BRD4 in this screening. Notably, miR-1293 also suppressed DNA repair pathways by directly suppressing the DNA repair genes APEX1 (apurinic-apyrimidinic endonuclease 1), RPA1 (replication protein A1), and POLD4 (DNA polymerase delta 4, accessory subunit). Concurrent suppression of BRD4 and these DNA repair genes synergistically inhibited tumor cell growth in vitro. Furthermore, administration of miR-1293 suppressed in vivo tumor growth in a xenograft mouse model. These results suggest that miR-1293 is a candidate for the development of miRNA-based cancer therapeutics.
Collapse
Affiliation(s)
- Yuki Takagawa
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuyuki Gen
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Tomoki Muramatsu
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kousuke Tanimoto
- Genome Laboratory, Medical Research Institute, TMDU, Tokyo, Japan
| | - Jun Inoue
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Johji Inazawa
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan; Bioresource Research Center, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
23
|
Huang W, Gu J, Tao T, Zhang J, Wang H, Fan Y. MiR-24-3p Inhibits the Progression of Pancreatic Ductal Adenocarcinoma Through LAMB3 Downregulation. Front Oncol 2020; 9:1499. [PMID: 32039003 PMCID: PMC6985431 DOI: 10.3389/fonc.2019.01499] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 12/16/2019] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with several genetic syndromes. However, the molecular mechanism underlying PDAC progression is still unknown. In this study, we showed that Laminin Subunit Beta 3 (LAMB3) was aberrantly overexpressed in PDAC and was closely associated with the overall survival rate of patients with PDAC. Functional studies demonstrated that LAMB3 played important roles in cell proliferation, the cell cycle, and invasion capacity. Using bioinformatics analysis, we determined that miR-24-3p was an upstream miRNA of LAMB3, and further experiments verified that miR-24-3p regulated LAMB3 expression in PDAC cells. A dual-luciferase reporter system demonstrated that miR-24-3p directly targeted the LAMB3 3'UTR, and FISH assay confirmed that miR-24-3p and LAMB3 mRNA mostly resided in cytoplasm, accounting for their post-translational regulation. Rescue assay demonstrated that miR-24-3p exerted its anti-cancer role by suppressing LAMB3 expression. Finally, by using a subcutaneous xenotransplanted tumor model, we demonstrated that miR-24-3p overexpression inhibited the proliferation of PDAC by suppressing LAMB3 expression in vivo. Collectively, our results provide evidence that the miR-24-3p/LAMB3 axis plays a vital role in the progression of PDAC and indicate that the miR-24-3p/LAMB3 axis may represent a novel therapeutic target for PDAC.
Collapse
Affiliation(s)
- Wenjie Huang
- Department of First Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Jianyou Gu
- Department of First Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tian Tao
- Department of First Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junfeng Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China.,Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Yingfang Fan
- Department of First Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Matboli M, Labib ME, Nasser HET, El-Tawdi AH, Habib EK, Ali-Labib R. Exosomal miR-1298 and lncRNA-RP11-583F2.2 Expression in Hepato-cellular Carcinoma. Curr Genomics 2020; 21:46-55. [PMID: 32655298 PMCID: PMC7324892 DOI: 10.2174/1389202920666191210111849] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/21/2022] Open
Abstract
AIM The aim of this study was to explore the expression of exosomal non-coding RNAs (ncRNAs) in the sera of patients with HCC versus control. METHODS Firstly, Bioinformatics analysis was conducted to retrieve ncRNAs specific to HCC (hsa-miRNA-1298 and lncRNA-RP11-583F2.2). Afterwards, extraction and characterization of exosomes were performed. We measured the expression of the chosen exosomal RNAs by reverse transcriptase quantitative real-time PCR in sera of 60 patients with HCC, 42 patients with chronic hepatitis C (CHC) infection and 18 healthy normal volunteers. RESULTS The exosomal ncRNAs [hsa-miRNA-1298, lncRNA-RP11-583F2.2] had better sensitivity and specificity than alpha-fetoprotein (AFP) in HCC diagnosis. CONCLUSION The exosomal hsa-miRNA-1298, lncRNA-RP11-583F2.2 can be potential biomarkers for HCC diagnosis.
Collapse
Affiliation(s)
- Marwa Matboli
- Address correspondence to this author at the Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbassia, 11381, Cairo, Egypt; Tel: 01005824962; E-mail:
| | | | | | | | | | | |
Collapse
|
25
|
Du Z, Wu J, Wang J, Liang Y, Zhang S, Shang Z, Zuo W. MicroRNA-1298 is downregulated in non-small cell lung cancer and suppresses tumor progression in tumor cells. Diagn Pathol 2019; 14:132. [PMID: 31801557 PMCID: PMC6894281 DOI: 10.1186/s13000-019-0911-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) have been reported to serve pivotal roles in tumorigenesis. This study sough to assess the expression and clinical significance of microRNA-1298 (miR-1298) in patients with non-small cell lung cancer (NSCLC), and explore the functional role of miR-1298 in tumorigenesis. METHODS One hundred and twenty-one NSCLC patients were recruited in this study. The expression of miR-1298 was estimated using quantitative real-time PCR. Kaplan-Meier survival curves and Cox regression analysis were used to evaluate the prognostic value of miR-1298. Gain- and loss-of-function experiments were preformed to explore the biological function of miR-1298 in NSCLC cells. RESULTS Expression levels of miR-1298 were downregulated in NSCLC tissues and cells compared with the corresponding normal controls. The decreased expression of miR-1298 was associated with patients' lymph node metastasis and TNM stage. The low expression of miR-1298 predicted poor overall survival and served as an independent prognostic indicator in NSCLC patients. According to the cell experiments, NSCLC cell proliferation, migration and invasion were inhibited by the overexpression of miR-1298. CONCLUSION All the data indicated that the downregulation of miR-1298 predicts poor prognosis of NSCLC, and the overexpression of miR-1298 in NSCLC cells leads to inhibited tumorigenesis. The aberrant miR-1298 may serve as a novel biomarker and therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Zhonghai Du
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China
| | - Jun Wu
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China
| | - Juan Wang
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China
| | - Yan Liang
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China.
| | - Sensen Zhang
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China
| | - Zhimei Shang
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China
| | - Wenchao Zuo
- Cancer center, Weifang Traditional Chinese Hospital, No. 1055 Weizhou Road, Weifang, 261041, Shandong, China
| |
Collapse
|
26
|
Li G, Sun L, Mu Z, Liu S, Qu H, Xie Q, Hu B. MicroRNA-1298-5p inhibits cell proliferation and the invasiveness of bladder cancer cells via down-regulation of connexin 43. Biochem Cell Biol 2019; 98:227-237. [PMID: 31600451 DOI: 10.1139/bcb-2019-0137] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNA (miR)-1298 is widely down-regulated in a variety of malignant tumors, which facilitates cell proliferation, invasiveness, and migration. However, the specific biological function of miR-1298 in bladder cancer (BC) is still unknown. Connexin 43 (Cx43) is often up-regulated in tumors. Identifying miRNAs that target Cx43 in the setting of BC will help to develop Cx43-based therapies for BC. In this study, the results demonstrated that the expression levels of miR-1298 and Cx43 were significantly down-regulated and up-regulated, respectively, in BC tissues. Overexpression of miR-1298 inhibited cell proliferation, migration, and invasiveness in two BC cell lines as determined using MTT assays, cell cycle assays, colony formation assays, Transwell assays, gelatin zymography, and Western blot. In addition, we found that miR-1298 decreased Cx43 expression by directly targeting the 3'-UTR. Further, we observed that the promotion of BC cell proliferation, migration, and invasiveness from Cx43 on could be partially attenuated by overexpressing miR-1298. Moreover, the protein expression of p-ERK was ameliorated after transfection with overexpressed-miR-1298. Knockdown of Cx43 reversed the promotion of cell migration and invasiveness due to decreased expression of miR-1298. All of the data from our study indicate that miR-1298 could be a diagnostic marker of BC and a potential therapeutic agent via inhibiting Cx43.
Collapse
Affiliation(s)
- Gang Li
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Longfeng Sun
- Department of Geriatric Cardiovascular Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Zhongyi Mu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Shibo Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Hongchen Qu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Qingpeng Xie
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| | - Bin Hu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, People's Republic of China
| |
Collapse
|
27
|
Hasanin AH, Matboli M, Seleem HS. Hesperidin suppressed hepatic precancerous lesions via modulation of exophagy in rats. J Cell Biochem 2019; 121:1295-1306. [PMID: 31489981 DOI: 10.1002/jcb.29363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 08/23/2019] [Indexed: 12/21/2022]
Abstract
The enormous cost of modern medicines warrants alternative strategies for the better management of hepatocellular carcinoma. Recently, exosomes have been shown to relay the oncogenic information through the horizontal transfer of RNAs between the cells. In this study, we modulated exosomal production and autophagy (exophagy) by the administration of hesperidin and evaluated its effect on the development of hepatic precancerous lesion (HPC) in rats. Diethylnitrosamine and 2-acetylaminofluorene were used in vivo to induce HPC in rats. Rats were allocated into five groups: naïve, HPC, and three hesperidin treated (50, 100, and 200 mg/kg/d; orally) for 4 consecutive days per week for 16 weeks. Liver tissues and blood samples were collected for histopathological, immunohistochemical, and transmission electron microscope examinations, liver function, alfa-fetoprotein level, and isolation of exosomal and autophagy RNAs. Hesperidin administration showed hepato-protective effects and improved the microscopic hepatic features with a decrease in glutathione S-transferase placental precancerous foci and the abundance of exosomes in liver tissues. Hesperidin improved liver function with a significant decrease in alfa-fetoprotein levels. Hesperidin dose-dependently decreased exosomal RAB11A messsenger RNA and long noncoding RNA-RP11-583F2.2 along with the increase in exosomal miR-1298, involved in the exophagy process. In conclusion, hesperidin likely suppresses liver carcinogenesis in rat model via the modulation of exosomal secretion and autophagy.
Collapse
Affiliation(s)
- Amany H Hasanin
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hanan S Seleem
- Department of Histology, Faculty of Medicine, Menoufia University, Cairo, Egypt.,Histology Department, Faculty of Medicine, Unaizah College of Medicine, Al Qassim University, Buraydah, KSA
| |
Collapse
|
28
|
Restoration of mutant K-Ras repressed miR-199b inhibits K-Ras mutant non-small cell lung cancer progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:165. [PMID: 30987652 PMCID: PMC6466664 DOI: 10.1186/s13046-019-1170-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/07/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND miRNAs play crucial role in the progression of K-Ras-mutated nonsmall cell lung cancer (NSCLC). However, most studies have focused on miRNAs that target K-Ras. Here, we investigated miRNAs regulated by mutant K-Ras and their functions. METHODS miRNAs regulated by mutant K-Ras were screened using miRNA arrays. miR-199b expression levels were measured by qRT-PCR. The protein expression levels were measured using Western blot and immunohistochemistry. The effects of miR-199b on NSCLC were examined both in vitro and in vivo by overexpressing or inhibiting miR-199b. DNA methylation was measured by bisulfite sequencing. RESULTS An inverse correlation was observed between K-Ras mutation status and miR-199b levels in NSCLC specimens and cell lines. The inhibition of miR-199b stimulated NSCLC growth and metastasis, while restoration of miR-199b suppressed K-Ras mutation-driven lung tumorigenesis as well as K-Ras-mutated NSCLC growth and metastasis. miR-199b inactivated ERK and Akt pathways by targeting K-Ras, KSR2, PIK3R1, Akt1, and Rheb1. Furthermore, we determined that mutant K-Ras inhibits miR-199b expression by increasing miR-199b promoter methylation. CONCLUSION Our findings suggest that mutant K-Ras plays an oncogenic role through downregulating miR-199b in NSCLC and that overexpression of miR-199b is a novel strategy for the treatment of K-Ras-mutated NSCLC.
Collapse
|
29
|
Exosomal miRNAs as Novel Pharmacodynamic Biomarkers for Cancer Chemopreventive Agent Early Stage Treatments in Chemically Induced Mouse Model of Lung Squamous Cell Carcinoma. Cancers (Basel) 2019; 11:cancers11040477. [PMID: 30987362 PMCID: PMC6520832 DOI: 10.3390/cancers11040477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 01/20/2023] Open
Abstract
Background: Chemopreventive agent (CPA) treatment is one of the main preventive options for lung cancer. However, few studies have been done on pharmacodynamic biomarkers of known CPAs for lung cancer. Materials and methods: In this study, we treated mouse models of lung squamous cell carcinoma with three different CPAs (MEK inhibitor: AZD6244, PI-3K inhibitor: XL-147 and glucocorticoid: Budesonide) and examined circulating exosomal miRNAs in the plasma of each mouse before and after treatment. Results: Compared to baselines, we found differentially expressed exosomal miRNAs after AZD6244 treatment (n = 8, FDR < 0.05; n = 55, raw p-values < 0.05), after XL-147 treatment (n = 4, FDR < 0.05; n = 26, raw p-values < 0.05) and after Budesonide treatment (n = 1, FDR < 0.05; n = 36, raw p-values < 0.05). In co-expression analysis, we found that modules of exosomal miRNAs reacted to CPA treatments differently. By variable selection, we identified 11, 9 and nine exosomal miRNAs as predictors for AZD6244, XL-147 and Budesonide treatment, respectively. Integrating all the results, we highlighted 4 miRNAs (mmu-miR-215-5p, mmu-miR-204-5p, mmu-miR-708-3p and mmu-miR-1298-5p) as the key for AZD6244 treatment, mmu-miR-23a-3p as key for XL-147 treatment, and mmu-miR-125a-5p and mmu-miR-16-5p as key for Budesonide treatment. Conclusions: This is the first study to use circulating exosomal miRNAs as pharmacodynamic biomarkers for CPA treatment in lung cancer.
Collapse
|
30
|
Zhang H, Pan YZ, Cheung M, Cao M, Yu C, Chen L, Zhan L, He ZW, Sun CY. LAMB3 mediates apoptotic, proliferative, invasive, and metastatic behaviors in pancreatic cancer by regulating the PI3K/Akt signaling pathway. Cell Death Dis 2019; 10:230. [PMID: 30850586 PMCID: PMC6408539 DOI: 10.1038/s41419-019-1320-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/08/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022]
Abstract
The poor prognosis of patients with pancreatic ductal adenocarcinoma (PDAC) is partially attributed to the invasive and metastatic behavior of this disease. Laminin subunit beta-3 (LAMB3) encodes one of the three subunits of LM-332, an extracellular matrix protein secreted by cultured human keratinocytes. In addition, LAMB3 is involved in the invasive and metastatic abilities of some types of cancer, including colon, pancreas, lung, cervix, stomach, and prostate cancer, but the role and mechanism of LAMB3 in PDAC have not been previously determined. Herein, we tentatively investigated the role of LAMB3 in the malignant biological behavior of PDAC. In this study, we demonstrated that LAMB3 is upregulated in PDAC. Inhibition of LAMB3 abrogated the tumorigenic outcomes of PI3K/Akt signaling pathway activation, including those involving cell cycle arrest, cell apoptosis, proliferation, invasion and migration in vitro, and tumor growth and liver metastasis in vivo. Our results showed that LAMB3 could mediate cell cycle arrest and apoptosis in PDAC cells and alter the proliferative, invasive, and metastatic behaviors of PDAC by regulating the PI3K/Akt signaling pathway. LAMB3 may be a novel therapeutic target for the treatment of PDAC in the future.
Collapse
Affiliation(s)
- Hong Zhang
- Guizhou Medical University, Guiyang, Guizhou, China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Yao-Zhen Pan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China
| | - May Cheung
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Mary Cao
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Chao Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ling Chen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Zhan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhi-Wei He
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China
| | - Cheng-Yi Sun
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Hepatobiliary-Pancreas-Spleen Surgery of Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
31
|
The Modulatory Role of MicroRNA-873 in the Progression of KRAS-Driven Cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:301-317. [PMID: 30654191 PMCID: PMC6348737 DOI: 10.1016/j.omtn.2018.11.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022]
Abstract
KRAS is one of the most frequently mutated proto-oncogenes in pancreatic ductal adenocarcinoma (PDAC) and aberrantly activated in triple-negative breast cancer (TNBC). A profound role of microRNAs (miRNAs) in the pathogenesis of human cancer is being uncovered, including in cancer therapy. Using in silico prediction algorithms, we identified miR-873 as a potential regulator of KRAS, and we investigated its role in PDAC and TNBC. We found that reduced miR-873 expression is associated with shorter patient survival in both cancers. miR-873 expression is significantly repressed in PDAC and TNBC cell lines and inversely correlated with KRAS levels. We demonstrate that miR-873 directly bound to the 3′ UTR of KRAS mRNA and suppressed its expression. Notably, restoring miR-873 expression induced apoptosis; recapitulated the effects of KRAS inhibition on cell proliferation, colony formation, and invasion; and suppressed the activity of ERK and PI3K/AKT, while overexpression of KRAS rescued the effects mediated by miR-873. Moreover, in vivo delivery of miR-873 nanoparticles inhibited KRAS expression and tumor growth in PDAC and TNBC tumor models. In conclusion, we provide the first evidence that miR-873 acts as a tumor suppressor by targeting KRAS and that miR-873-based gene therapy may be a therapeutic strategy in PDAC and TNBC.
Collapse
|
32
|
Carpenter PM, Ziogas A, Markham EM, Cantillep AS, Yan R, Anton-Culver H. Laminin 332 expression and prognosis in breast cancer. Hum Pathol 2018; 82:289-296. [PMID: 30125583 PMCID: PMC6289632 DOI: 10.1016/j.humpath.2018.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
The purpose of this study was to determine the distribution of and potential significance of laminin 332 (LM332) in breast cancer. Specimens from a population-based cohort (N = 297) from 1994 to 1995 were stained for estrogen receptor (ER), progesterone receptor (PgR), HER2 and the LM332 β3 chain. Seventy-five tumors were LM332-positive and 222 were negative. LM332 β3 stained 16.0% of ER and/or PgR-positive tumors and 73.2% of triple-negative breast cancers (TNBC). Immunoblotting revealed LM332 in TNBC and HER2-positive samples, but not in an ER-positive breast carcinoma or a phyllodes tumor. After 20 years, 172 patients were alive, 43 had died of breast cancer and 82 of other causes. Patients with LM332-positive tumors had significantly worse 5 (P < .0001) and 10-year (P < .05) overall and breast cancer specific survival. Among patients with LM332 β3-expressing and ER/PgR-negative carcinomas, 10-year survival was significantly reduced (P < .0450). In a multivariate analysis LM332-positive patients had significant hazard ratios of 3.9 with 95% confidence intervals (CI) of 2.0-7.7 and 2.2 with 95% CI of 1.3-3.8 for 5 and 10-year overall survival, respectively. Because tumor cell motility is required for metastasis, the effect of LM332 on MDA-MB-231 migration was determined using siRNA. Knockdown of LM332-specific β3 and γ2 chains reduced motility without affecting viability. Our observation that LM332 in breast carcinoma is associated with decreased survival provides evidence that LM332 may have a role in the aggressive phenotype of some breast cancers.
Collapse
Affiliation(s)
- Philip M Carpenter
- Department of Pathology, Keck School of Medicine, the University of Southern California; Los Angeles, CA, USA; Department of Pathology, University of California, Orange, CA, USA.
| | - Argyrios Ziogas
- Department of Epidemiology, University of California, Irvine, CA 92697-7550, USA.
| | - Emma M Markham
- Department of Pathology, University of California, Orange, CA, USA; Department of Epidemiology, University of California, Irvine, CA 92697-7550, USA.
| | | | - Rui Yan
- Department of Pathology, Keck School of Medicine, the University of Southern California; Los Angeles, CA, USA.
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, CA 92697-7550, USA
| |
Collapse
|
33
|
Saeed O, Lopez-Beltran A, Fisher KW, Scarpelli M, Montironi R, Cimadamore A, Massari F, Santoni M, Cheng L. RAS genes in colorectal carcinoma: pathogenesis, testing guidelines and treatment implications. J Clin Pathol 2018; 72:135-139. [PMID: 30425122 DOI: 10.1136/jclinpath-2018-205471] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022]
Abstract
The RAS family is among the most commonly mutated genes in all human malignancies including colon cancer. In normal cells, RAS proteins act as a link in the intracellular signal transduction initiated by binding of growth factors to cell membrane receptors mediating cell survival. RAS isoproteins have great morphological similarities, but despite that, they are thought to have different functions in different tissues. RAS mutations, as supported by several studies including animal models, have a role in the development and progression of colorectal cancer. The detection of RAS mutations in patients with colorectal carcinoma, specifically KRAS and NRAS, has significant clinical implications. It is currently recommended that patients with colon cancer who are considered for antiepidermal growth factor receptor monoclonal antibodies, get RAS mutation testing since only those with wildtype-RAS genes benefit from such treatment. Despite decades of research, there is currently no effective and safe treatment that directly targets RAS-mutated neoplasms. Multiple therapeutic approaches directed against RAS mutations are currently experimental, including a promising immunotherapy study using T-cells in patients with metastatic colon cancer.
Collapse
Affiliation(s)
- Omer Saeed
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | | | - Kurt W Fisher
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, USA
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| |
Collapse
|
34
|
Zhang YS, Chen T, Cai YJ, Dong J, Bai F, Gao X, Tian L, Duan N, Liu D. MicroRNA-647 promotes the therapeutic effectiveness of argon-helium cryoablation and inhibits cell proliferation through targeting TRAF2 via the NF-κB signaling pathway in non-small cell lung cancer. Onco Targets Ther 2018; 11:6777-6784. [PMID: 30349310 PMCID: PMC6188019 DOI: 10.2147/ott.s159337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background MicroRNA-647 (miR-647) has been reported to repress cell tumorigenic phenotype, while the function of miR-647 in non-small cell lung cancer was obscure. Methods The effect of miR-647 and TRAF2 on A549 and H1299 cells was explored through Methyl thiazolyl tetrazolium (MTT) assay, colony formation and cell cycle assays. Luciferase reporter assays, reverse transcription quantitative PCR (RT-qPCR) and Western blot assay were carried out to determine that TRAF2 is directly regulated by miR-647. The effect of miR-647/TRAF2 axis on p65 protein level in nucleus or total was detected by Western blot assay. Results Here, we found that miR-647 was high expression in tumor that under argon-helium cryoablation treatment in contrast to the tumor under non argon-helium cryoablation treatment and inhibited cell proliferation of A549 and H1299 cells by inducing G1-S transition. TRAF2 was confirmed as a target of miR-647. TRAF2 overexpression partially rescued the suppressive function of miR-647 in A549 and H1299 cells. Moreover, we found that miR-647 repressed lung carcinogenesis by attenuating NF-κB pathway. Conclusion In all, our study demonstrates that miR-647 functions as tumor suppressor via targeting and down-regulating the expression of TRAF2 and NF-κB signaling pathway in non-small cell lung cancer.
Collapse
Affiliation(s)
- Yun-Song Zhang
- Department of Thoracic Surgery, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China,
| | - Tianzi Chen
- Department of Thoracic Surgery, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China,
| | - Ying Jiu Cai
- Department of Thoracic Surgery, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China,
| | - Jianlin Dong
- Department of Thoracic Surgery, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China,
| | - Fang Bai
- Department of Thoracic Surgery, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China,
| | - Xiaojun Gao
- Department of Thoracic Surgery, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China,
| | - Li Tian
- Department of Clinical Pharmacy, Tianjin Medical University, Tianjin, People's Republic of China
| | - Naiying Duan
- Department of Clinical Pharmacy, Tianjin Medical University, Tianjin, People's Republic of China
| | - Dan Liu
- Key Specialist Office, Tianjin University of TCM Affiliated Wuqing Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
35
|
Liu K, Guo J, Liu K, Fan P, Zeng Y, Xu C, Zhong J, Li Q, Zhou Y. Integrative analysis reveals distinct subtypes with therapeutic implications in KRAS-mutant lung adenocarcinoma. EBioMedicine 2018; 36:196-208. [PMID: 30268834 PMCID: PMC6197714 DOI: 10.1016/j.ebiom.2018.09.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND KRAS-mutant lung adenocarcinomas (LUADs) are heterogeneous and frequently occur in smokers. The heterogeneity of KRAS-mutant LUAD has been an obstacle for the drug discovery. METHODS We integrated multiplatform datatypes and identified two corresponding subtypes in the patients and cell lines. We further characterized the features of these two subtypes and performed drug screening to identify subtype-specific drugs. Finally, we used the defining features of the KRAS subtypes for drug sensitivity prediction. FINDINGS Patient-Subtype 1 (PS1) was characterized by increased smoking-related mutational signature activity, a low tumor-infiltrating lymphocyte (TIL)-associating score and STK11/KEAP1 co-mutations. Patient-Subtype 2 (PS2) was characterized by an increased smoking-related methylation signature activity, a high TIL-associating score and increased KRAS dependency. The cell line subtypes faithfully recapitulated all the patients' features. Drug screening of the two cell line subtypes yielded several potential candidates, such as cytarabine and enzastaurin for Cell-line-Subtype 1 (CS1) and a BTK inhibitor QL-XII-61 for Cell-line-Subtype 2 (CS2). The defining features, such as smoking-related methylation signature, were significantly associated with the sensitivity to several drugs. INTERPRETATION The heterogeneity of KRAS-mutant LUAD is associated with smoking-related genomic and epigenomic aberration along with other features such as immunogenicity, KRAS dependency and STK11/KEAP1 co-mutations. These features might be used as biomarkers for drug sensitivity prediction. FUND: This research was funded by the Young Scientists Fund of the National Natural Science Foundation of China, the Natural Science Foundation of Fujian Province, China and the Education and Research Foundation for Young Scholars of Education Department of Fujian Province, China.
Collapse
Affiliation(s)
- Ke Liu
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Jintao Guo
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Kuai Liu
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Peiyang Fan
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Yuanyuan Zeng
- BGI-Shenzhen, Bei Shan Industrial Zone, Yantian District, Shenzhen, Guangdong Province 518083, China
| | - Chaoqun Xu
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Jiaxin Zhong
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Qiyuan Li
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China.
| | - Ying Zhou
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen 361102, China; Center for Biomedical Big Data Research, Medical College of Xiamen University, Xiamen 361102, China.
| |
Collapse
|
36
|
Naser R, Aldehaiman A, Díaz-Galicia E, Arold ST. Endogenous Control Mechanisms of FAK and PYK2 and Their Relevance to Cancer Development. Cancers (Basel) 2018; 10:E196. [PMID: 29891810 PMCID: PMC6025627 DOI: 10.3390/cancers10060196] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/31/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Focal adhesion kinase (FAK) and its close paralogue, proline-rich tyrosine kinase 2 (PYK2), are key regulators of aggressive spreading and metastasis of cancer cells. While targeted small-molecule inhibitors of FAK and PYK2 have been found to have promising antitumor activity, their clinical long-term efficacy may be undermined by the strong capacity of cancer cells to evade anti-kinase drugs. In healthy cells, the expression and/or function of FAK and PYK2 is tightly controlled via modulation of gene expression, competing alternatively spliced forms, non-coding RNAs, and proteins that directly or indirectly affect kinase activation or protein stability. The molecular factors involved in this control are frequently deregulated in cancer cells. Here, we review the endogenous mechanisms controlling FAK and PYK2, and with particular focus on how these mechanisms could inspire or improve anticancer therapies.
Collapse
Affiliation(s)
- Rayan Naser
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Abdullah Aldehaiman
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Escarlet Díaz-Galicia
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| |
Collapse
|
37
|
Ruibin W, Zheng X, Chen J, Zhang X, Yang X, Lin Y. Micro RNA-1298 opposes the effects of chronic oxidative stress on human trabecular meshwork cells via targeting on EIF4E3. Biomed Pharmacother 2018; 100:349-357. [PMID: 29453044 DOI: 10.1016/j.biopha.2018.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/23/2018] [Accepted: 02/02/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the effect and potential mechanism of miR-1298 in the progression of human trabecular meshwork (HTM) cells. MATERIAL AND METHODS Expression of miR-1298 was assessed by quantitative real time PCR (qRT-PCR), as well as in HTM-1 and HTM-2 cells. Mature miR-1298 mimic, miR-1298 inhibitor, and si-EIF4E3 and their corresponding controls were transfected into HTM-1 and HTM-2 to obtain stable HTM cells. Luciferase reporter assay was used to verify regulation between miR-1298 and EIF4E3. Cytotoxicity and Oxidative damage were assessed using commercial kits, and apoptosis was determined using flow cytometry. ECM and apoptosis related factors were determined using qRT-PCR and western blotting, as well as the pathway related factors. RESULTS The expression of miR-1298 was significantly decreased both in glaucoma and HTM cells. MiR-1298 mimic could significantly inhibit the increase of cytotoxicity, apoptosis, accumulation of carbonylated proteins and ECM induced by COS, but miR-1298 inhibitor could obviously promote the increase effects caused by COS in HTM cells. EIF4E3 was a downstream target of miR-1298. Sliced EIF4E3 could significantly inhibit the increase effects induced miR-1298 inhibitor in HTM cells under COS. The expression levels of TGF-β2 and Smad4 were significantly increased, and Wnt3a and β-cantenin were obviously decreased under COS, and miR-1298 inhibitor could markedly promote this increase effect, while sliced EIF4E3 could reverse the effect of miR-1298 under COS. CONCLUSIONS miR-1298 could protect HTM cells to against damage caused by COS via inhibiting TGF-β2/Smad4 pathway and activating canonical Wnt pathway.
Collapse
Affiliation(s)
- Wu Ruibin
- Department of Ophthalmolog, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515000, China.
| | - Xiaowei Zheng
- Department of Ophthalmolog, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Jiaying Chen
- Department of Ophthalmolog, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Xinyi Zhang
- Department of Ophthalmolog, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Xiayin Yang
- Department of Ophthalmolog, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Yuxian Lin
- Department of Ophthalmolog, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515000, China
| |
Collapse
|
38
|
Porru M, Pompili L, Caruso C, Biroccio A, Leonetti C. Targeting KRAS in metastatic colorectal cancer: current strategies and emerging opportunities. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018. [PMID: 29534749 PMCID: PMC5850913 DOI: 10.1186/s13046-018-0719-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Developing drugs that target KRAS, the most frequently mutated oncogene in cancer, has not been successful despite much concerted efforts dedicated towards it in the last thirty years. Considering the key role this driver oncogene plays, the pharmacological drugging of KRAS remains a key challenge for cancer research. In this review, we highlight the emerging experimental strategies for blocking KRAS function and signaling and its direct targeting. We also report on the results in this field of research produced by our group.
Collapse
Affiliation(s)
- Manuela Porru
- UOSD SAFU, Regina Elena National Cancer Institute, Rome, Italy
| | - Luca Pompili
- UOSD SAFU, Regina Elena National Cancer Institute, Rome, Italy.,University of Tuscia, Viterbo, Italy
| | | | - Annamaria Biroccio
- Oncogenomic and Epigenetic Unit, Regina Elena National Cancer Institute, Rome, Italy.
| | - Carlo Leonetti
- UOSD SAFU, Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
39
|
Wu P, Liu JL, Pei SM, Wu CP, Yang K, Wang SP, Wu S. Integrated genomic analysis identifies clinically relevant subtypes of renal clear cell carcinoma. BMC Cancer 2018. [PMID: 29534679 PMCID: PMC5851245 DOI: 10.1186/s12885-018-4176-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Renal cell carcinoma (RCC) account for over 80% of renal malignancies. The most common type of RCC can be classified into three subtypes including clear cell, papillary and chromophobe. ccRCC (the Clear Cell Renal Cell Carcinoma) is the most frequent form and shows variations in genetics and behavior. To improve accuracy and personalized care and increase the cure rate of cancer, molecular typing for individuals is necessary. Methods We adopted the genome, transcriptome and methylation HMK450 data of ccRCC in The Cancer Genome Atlas Network in this research. Consensus Clustering algorithm was used to cluster the expression data and three subtypes were found. To further validate our results, we analyzed an independent data set and arrived at a consistent conclusion. Next, we characterized the subtype by unifying genomic and clinical dimensions of ccRCC molecular stratification. We also implemented GSEA between the malignant subtype and the other subtypes to explore latent pathway varieties and WGCNA to discover intratumoral gene interaction network. Moreover, the epigenetic state changes between subgroups on methylation data are discovered and Kaplan-Meier survival analysis was performed to delve the relation between specific genes and prognosis. Results We found a subtype of poor prognosis in clear cell renal cell carcinoma, which is abnormally upregulated in focal adhesions and cytoskeleton related pathways, and the expression of core genes in the pathways are negatively correlated with patient outcomes. Conclusions Our work of classification schema could provide an applicable framework of molecular typing to ccRCC patients which has implications to influence treatment decisions, judge biological mechanisms involved in ccRCC tumor progression, and potential future drug discovery. Electronic supplementary material The online version of this article (10.1186/s12885-018-4176-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peng Wu
- The Affiliated Luohu Hospital of Shenzhen University, Department of Urological Surgery, Shenzhen University, Shenzhen, 518000, China.,Shenzhen Following Precision Medical Institute, Shenzhen Luohu Hospital Group, Shenzhen, 518000, China
| | - Jia-Li Liu
- Shenzhen Second People'Hospital, 1st affiliated hospital of ShenZhen University, Shenzhen, 518037, China
| | - Shi-Mei Pei
- Shenzhen Following Precision Medical Institute, Shenzhen Luohu Hospital Group, Shenzhen, 518000, China.,College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chang-Peng Wu
- Shenzhen Second People'Hospital, 1st affiliated hospital of ShenZhen University, Shenzhen, 518037, China
| | - Kai Yang
- The Affiliated Luohu Hospital of Shenzhen University, Department of Urological Surgery, Shenzhen University, Shenzhen, 518000, China.,Shenzhen Following Precision Medical Institute, Shenzhen Luohu Hospital Group, Shenzhen, 518000, China
| | - Shu-Peng Wang
- The Affiliated Luohu Hospital of Shenzhen University, Department of Urological Surgery, Shenzhen University, Shenzhen, 518000, China.,Shenzhen Following Precision Medical Institute, Shenzhen Luohu Hospital Group, Shenzhen, 518000, China
| | - Song Wu
- The Affiliated Luohu Hospital of Shenzhen University, Department of Urological Surgery, Shenzhen University, Shenzhen, 518000, China. .,Shenzhen Following Precision Medical Institute, Shenzhen Luohu Hospital Group, Shenzhen, 518000, China.
| |
Collapse
|
40
|
Román M, Baraibar I, López I, Nadal E, Rolfo C, Vicent S, Gil-Bazo I. KRAS oncogene in non-small cell lung cancer: clinical perspectives on the treatment of an old target. Mol Cancer 2018; 17:33. [PMID: 29455666 PMCID: PMC5817724 DOI: 10.1186/s12943-018-0789-x] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Lung neoplasms are the leading cause of death by cancer worldwide. Non-small cell lung cancer (NSCLC) constitutes more than 80% of all lung malignancies and the majority of patients present advanced disease at onset. However, in the last decade, multiple oncogenic driver alterations have been discovered and each of them represents a potential therapeutic target. Although KRAS mutations are the most frequently oncogene aberrations in lung adenocarcinoma patients, effective therapies targeting KRAS have yet to be developed. Moreover, the role of KRAS oncogene in NSCLC remains unclear and its predictive and prognostic impact remains controversial. The study of the underlying biology of KRAS in NSCLC patients could help to determine potential candidates to evaluate novel targeted agents and combinations that may allow a tailored treatment for these patients. The aim of this review is to update the current knowledge about KRAS-mutated lung adenocarcinoma, including a historical overview, the biology of the molecular pathways involved, the clinical relevance of KRAS mutations as a prognostic and predictive marker and the potential therapeutic approaches for a personalized treatment of KRAS-mutated NSCLC patients.
Collapse
Affiliation(s)
- Marta Román
- Department of Oncology, Clínica Universidad de Navarra, 31008, Pamplona, Spain.,Program of Solid Tumors and Biomarkers, Center for Applied Medical Research, Pamplona, Spain
| | - Iosune Baraibar
- Department of Oncology, Clínica Universidad de Navarra, 31008, Pamplona, Spain.,Program of Solid Tumors and Biomarkers, Center for Applied Medical Research, Pamplona, Spain
| | - Inés López
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research, Pamplona, Spain
| | - Ernest Nadal
- Thoracic Oncology Unit, Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Christian Rolfo
- Phase I-Early Clinical Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Edegem, Belgium
| | - Silvestre Vicent
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Gil-Bazo
- Department of Oncology, Clínica Universidad de Navarra, 31008, Pamplona, Spain. .,Program of Solid Tumors and Biomarkers, Center for Applied Medical Research, Pamplona, Spain. .,Navarra Health Research Institute (IDISNA), Pamplona, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
41
|
Molecular genetics and cellular events of K-Ras-driven tumorigenesis. Oncogene 2017; 37:839-846. [PMID: 29059163 PMCID: PMC5817384 DOI: 10.1038/onc.2017.377] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/11/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023]
Abstract
Cellular transformation and the accumulation of genomic instability are the two key events required for tumorigenesis. K-Ras (Kirsten-rat sarcoma viral oncogene homolog) is a prominent oncogene that has been proven to drive tumorigenesis. K-Ras also modulates numerous genetic regulatory mechanisms and forms a large tumorigenesis network. In this review, we track the genetic aspects of K-Ras signaling networks and assemble the sequence of cellular events that constitute the tumorigenesis process, such as regulation of K-Ras expression (which is influenced by miRNA, small nucleolar RNA and lncRNA), activation of K-Ras (mutations), generation of reactive oxygen species (ROS), induction of DNA damage and apoptosis, induction of DNA damage repair pathways and ROS detoxification systems, cellular transformation after apoptosis by the blebbishield emergency program and the accumulation of genomic/chromosomal instability that leads to tumorigenesis.
Collapse
|
42
|
García-Casas A, García-Olmo DC, García-Olmo D. Further the liquid biopsy: Gathering pieces of the puzzle of genometastasis theory. World J Clin Oncol 2017; 8:378-388. [PMID: 29067274 PMCID: PMC5638713 DOI: 10.5306/wjco.v8.i5.378] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/03/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
Metastasis is the major cause of mortality in cancer disease and still constitutes one of the most controversial mechanism, not yet fully understood. What is almost beyond doubt is that circulatory system is crucial for cancer propagation. Regarding this system, much attention has been recently paid to liquid biopsy. This technique is aimed to detect circulating tumor cells (CTCs) and circulating nucleic acids so it can be used as a tool for diagnostic, prognostic and follow-up of patients. Whereas CTCs tend to be scarce in serum and plasma from cancer patient, abundant circulating nucleic acids can be detected in the same location. This fact, together with the genetic origin of cancer, stands out the relevance of circulating nucleic acids and shed light into the role of nucleic acids as drivers of metastasis, a recently discovered phenomenon called Genometastasis. This innovative theory supports the transfer of oncogenes from cancer cells to normal and susceptible cells located in distant target organs through circulatory system. What is more, many biological processes haven been described to deliver and secrete circulating nucleic acids into the circulation which can allow such horizontal transfer of oncogenes. In this review, we focus not only on these mechanisms but also we demonstrate its putative role in cancer propagation and give insights about possible therapeutic strategies based on this theory. Our objective is to demonstrate how findings about cell-to-cell communications and previous results can agree with this unprecedented theory.
Collapse
Affiliation(s)
| | - Dolores C García-Olmo
- Centre de Recerca Experimental Biomèdica Aplicada(CREBA), IRBLLEIDA, 25138 Lleida, Spain
| | - Damián García-Olmo
- Department of Surgery, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria, Fundación Jiménez Díaz, 28050 Madrid, Spain
| |
Collapse
|
43
|
Yau EH, Kummetha IR, Lichinchi G, Tang R, Zhang Y, Rana TM. Genome-Wide CRISPR Screen for Essential Cell Growth Mediators in Mutant KRAS Colorectal Cancers. Cancer Res 2017; 77:6330-6339. [PMID: 28954733 DOI: 10.1158/0008-5472.can-17-2043] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/22/2022]
Abstract
Targeting mutant KRAS signaling pathways continues to attract attention as a therapeutic strategy for KRAS-driven tumors. In this study, we exploited the power of the CRISPR-Cas9 system to identify genes affecting the tumor xenograft growth of human mutant KRAS (KRASMUT) colorectal cancers. Using pooled lentiviral single-guide RNA libraries, we conducted a genome-wide loss-of-function genetic screen in an isogenic pair of human colorectal cancer cell lines harboring mutant or wild-type KRAS. The screen identified novel and established synthetic enhancers or synthetic lethals for KRASMUT colorectal cancer, including targetable metabolic genes. Notably, genetic disruption or pharmacologic inhibition of the metabolic enzymes NAD kinase or ketohexokinase was growth inhibitory in vivo In addition, the chromatin remodeling protein INO80C was identified as a novel tumor suppressor in KRASMUT colorectal and pancreatic tumor xenografts. Our findings define a novel targetable set of therapeutic targets for KRASMUT tumors. Cancer Res; 77(22); 6330-9. ©2017 AACR.
Collapse
Affiliation(s)
- Edwin H Yau
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California.,Division of Hematology-Oncology, Department of Internal Medicine, University of California San Diego School of Medicine, La Jolla, California.,Solid Tumor Therapeutics Program, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Indrasena Reddy Kummetha
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Gianluigi Lichinchi
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Rachel Tang
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Yunlin Zhang
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Tariq M Rana
- Department of Pediatrics and Institute for Genomic Medicine, University of California San Diego School of Medicine, La Jolla, California. .,Solid Tumor Therapeutics Program, Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
44
|
Schultz DJ, Muluhngwi P, Alizadeh-Rad N, Green MA, Rouchka EC, Waigel SJ, Klinge CM. Genome-wide miRNA response to anacardic acid in breast cancer cells. PLoS One 2017; 12:e0184471. [PMID: 28886127 PMCID: PMC5590942 DOI: 10.1371/journal.pone.0184471] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs are biomarkers and potential therapeutic targets for breast cancer. Anacardic acid (AnAc) is a dietary phenolic lipid that inhibits both MCF-7 estrogen receptor α (ERα) positive and MDA-MB-231 triple negative breast cancer (TNBC) cell proliferation with IC50s of 13.5 and 35 μM, respectively. To identify potential mediators of AnAc action in breast cancer, we profiled the genome-wide microRNA transcriptome (microRNAome) in these two cell lines altered by the AnAc 24:1n5 congener. Whole genome expression profiling (RNA-seq) and subsequent network analysis in MetaCore Gene Ontology (GO) algorithm was used to characterize the biological pathways altered by AnAc. In MCF-7 cells, 69 AnAc-responsive miRNAs were identified, e.g., increased let-7a and reduced miR-584. Fewer, i.e., 37 AnAc-responsive miRNAs were identified in MDA-MB-231 cells, e.g., decreased miR-23b and increased miR-1257. Only two miRNAs were increased by AnAc in both cell lines: miR-612 and miR-20b; however, opposite miRNA arm preference was noted: miR-20b-3p and miR-20b-5p were upregulated in MCF-7 and MDA-MB-231, respectively. miR-20b-5p target EFNB2 transcript levels were reduced by AnAc in MDA-MB-231 cells. AnAc reduced miR-378g that targets VIM (vimentin) and VIM mRNA transcript expression was increased in AnAc-treated MCF-7 cells, suggesting a reciprocal relationship. The top three enriched GO terms for AnAc-treated MCF-7 cells were B cell receptor signaling pathway and ribosomal large subunit biogenesis and S-adenosylmethionine metabolic process for AnAc-treated MDA-MB-231 cells. The pathways modulated by these AnAc-regulated miRNAs suggest that key nodal molecules, e.g., Cyclin D1, MYC, c-FOS, PPARγ, and SIN3, are targets of AnAc activity.
Collapse
Affiliation(s)
- David J. Schultz
- Department of Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Penn Muluhngwi
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Negin Alizadeh-Rad
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Madelyn A. Green
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Eric C. Rouchka
- Bioinformatics and Biomedical Computing Laboratory, Department of Computer Engineering and Computer Science, Louisville, Kentucky, United States of America
| | - Sabine J. Waigel
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Carolyn M. Klinge
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| |
Collapse
|
45
|
Xu M, Qin S, Cao F, Ding S, Li M. MicroRNA-379 inhibits metastasis and epithelial-mesenchymal transition via targeting FAK/AKT signaling in gastric cancer. Int J Oncol 2017; 51:867-876. [DOI: 10.3892/ijo.2017.4072] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 07/04/2017] [Indexed: 11/05/2022] Open
|
46
|
Vallejo A, Valencia K, Vicent S. All for one and FOSL1 for all: FOSL1 at the crossroads of lung and pancreatic cancer driven by mutant KRAS. Mol Cell Oncol 2017; 4:e1314239. [PMID: 28616588 DOI: 10.1080/23723556.2017.1314239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 10/19/2022]
Abstract
KRAS proto-oncogene, GTPase (KRAS) remains refractory to current therapies. We devised an integrative cross-tumor approach to expose common core elements up-regulated in mutant KRAS cancers that could provide new treatment opportunities. This approach identified FOSL1 (Fos-like antigen 1) as a clinically and functionally relevant gene in mutant KRAS-driven lung and pancreatic cancers, and unveiled downstream transcriptional targets amenable to pharmacological inhibition.
Collapse
Affiliation(s)
- Adrian Vallejo
- Program in Solid Tumors and Biomarkers, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain.,Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Karmele Valencia
- Program in Solid Tumors and Biomarkers, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | - Silvestre Vicent
- Program in Solid Tumors and Biomarkers, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain.,Department of Histology and Pathology, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|