1
|
Gao XQ, Li HL, Wang M, Yang CT, Su R, Shao LH. Kaempferol inhibited invasion and metastasis of gastric cancer cells by targeting AKT/GSK3β pathway based on network pharmacology and molecular docking. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:421-441. [PMID: 39132822 DOI: 10.1080/10286020.2024.2387756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
This study aims to explore the mechanisms of the inhibitory effect of kaempferol on the invasion and metastasis of gastric cancer (GC) cells through network pharmacology prediction and experimental verification. It identifies core targets via PPI network analysis and finds that kaempferol binds to these targets well. In vitro experiments showed that kaempferol could inhibit the proliferation, colony formation, migration and invasion of GC cells. Western blotting indicated kaempferol may reduce AKT and GSK3β phosphorylation, leading to lower expression of invasion-related genes SRC, MMP9, CXCR4, KDR, and MMP2. Overall, kaempferol may prevent migration and invasion of GC cells via the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Xia-Qing Gao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Hai-Long Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Department of Geriatrics, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Meng Wang
- Department of Geriatrics, Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, China
- Research Center of Traditional Chinese Medicine, Gansu Province, Lanzhou 730000, China
| | - Chun-Ting Yang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Rong Su
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Li-Hua Shao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| |
Collapse
|
2
|
Liang JE, Bao BW, He XH, Lu WQ, Liu Y, Wang J, Qu XJ, Li DY, Che XF. LOXL1 promotes gastric cancer progression by β-catenin-cyclinD mediated proliferation. Exp Cell Res 2024; 443:114331. [PMID: 39547354 DOI: 10.1016/j.yexcr.2024.114331] [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: 05/28/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Although much progress has been made in chemotherapy or target therapy for advanced gastric cancer, the prognosis is still poor. It is necessary to screen biomarkers for early diagnosis and prognosis prediction. However, the prognostic value of LOX family in gastric cancer and the underlying molecular mechanisms for promoting the progression of gastric cancer remains unclear. Among five members of LOX family, LOXL1 was the unique independent prognostic risk factor. The nomogram established based on the expression of LOXL1 and other clinical parameters could predict the overall survival rate of gastric cancer. Knockdown (KD) of LOXL1 decreased cell proliferation and led to G1 phase arrest in gastric cells. According to GSEA analysis that LOXL1 was positively correlated with the WNT signaling pathway, in vitro experiment proved that LOXL1-KD reduced the phosphorylation level of β-catenin and the expression of the downstream G1 phase checkpoint CCND1. In conclusion, LOXL1 has been identified as a potential risk prognostic biomarker for gastric cancer by promoting gastric cancer proliferation via WNT/β-catenin/cyclinD1 pathway.
Collapse
Affiliation(s)
- Jin-E Liang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Bo-Wen Bao
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Xue-Hua He
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China; Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wen-Qing Lu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yang Liu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Jin Wang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiu-Juan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Dong-Yang Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
| | - Xiao-Fang Che
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
3
|
Park SH, Choi SH, Park HY, Ko J, Yoon JS. Role of Lysyl Oxidase-Like Protein 3 in the Pathogenesis of Graves' Orbitopathy in Orbital Fibroblasts. Invest Ophthalmol Vis Sci 2024; 65:33. [PMID: 39546293 DOI: 10.1167/iovs.65.13.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
Purpose The lysyl oxidase (LOX) family has been implicated in the pathogenesis of diseases caused by inflammation and fibrosis. Therefore, we aimed to examine the role of lysyl oxidase-like protein 3 (LOXL3) in Graves' orbitopathy (GO) pathogenesis and its potential as a treatment target. Methods Quantitative real-time polymerase chain reaction compared the transcript levels of the five LOX family subtypes in orbital tissue explants obtained from patients with GO (n = 18) and healthy controls (n = 15). The effects of LOXL3 inhibition on interleukin (IL)-1β-induced proinflammatory cytokines, transforming growth factor (TGF)-β-induced profibrotic proteins, intracellular signaling molecules, and adipogenic markers were evaluated using Western blotting. Adipogenic differentiation was identified using Oil Red O staining. Results LOX and LOXL3 transcript levels were high in GO tissues. Stimulation with IL-1β, TGF-β, and insulin-like growth factor-1 significantly increased LOXL3 messenger RNA expression in GO fibroblasts. Furthermore, silencing LOXL3 attenuated the IL-1β-induced production of proinflammatory cytokines (IL-6, IL-8, and intercellular adhesion molecule-1) and TGF-β-induced production of profibrotic proteins (fibronectin, collagen 1α, and alpha-smooth muscle actin). It also reduced the IL-1β or TGF-β-induced expression of phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells, protein kinase B, extracellular signal-regulated kinase, and c-Jun N-terminal kinase. Additionally, LOXL3 silencing suppressed adipocyte differentiation and the expression of adipogenic transcription factors (leptin, AP-2, peroxisome proliferator-activated receptor gamma, and CCAAT/enhancer-binding protein). Conclusions LOXL3 is crucial in GO pathogenesis. LOXL3 inhibition reduced inflammatory cytokine production, fibrotic protein expression, and fibroblast differentiation into adipocytes. This study highlights LOXL3 as a potential therapeutic target for GO.
Collapse
Affiliation(s)
| | - Soo Hyun Choi
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Young Park
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSang Ko
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Ozmen E, Demir TD, Ozcan G. Cancer-associated fibroblasts: protagonists of the tumor microenvironment in gastric cancer. Front Mol Biosci 2024; 11:1340124. [PMID: 38562556 PMCID: PMC10982390 DOI: 10.3389/fmolb.2024.1340124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
Enhanced knowledge of the interaction of cancer cells with their environment elucidated the critical role of tumor microenvironment in tumor progression and chemoresistance. Cancer-associated fibroblasts act as the protagonists of the tumor microenvironment, fostering the metastasis, stemness, and chemoresistance of cancer cells and attenuating the anti-cancer immune responses. Gastric cancer is one of the most aggressive cancers in the clinic, refractory to anti-cancer therapies. Growing evidence indicates that cancer-associated fibroblasts are the most prominent risk factors for a poor tumor immune microenvironment and dismal prognosis in gastric cancer. Therefore, targeting cancer-associated fibroblasts may be central to surpassing resistance to conventional chemotherapeutics, molecular-targeted agents, and immunotherapies, improving survival in gastric cancer. However, the heterogeneity in cancer-associated fibroblasts may complicate the development of cancer-associated fibroblast targeting approaches. Although single-cell sequencing studies started dissecting the heterogeneity of cancer-associated fibroblasts, the research community should still answer these questions: "What makes a cancer-associated fibroblast protumorigenic?"; "How do the intracellular signaling and the secretome of different cancer-associated fibroblast subpopulations differ from each other?"; and "Which cancer-associated fibroblast subtypes predominate specific cancer types?". Unveiling these questions can pave the way for discovering efficient cancer-associated fibroblast targeting strategies. Here, we review current knowledge and perspectives on these questions, focusing on how CAFs induce aggressiveness and therapy resistance in gastric cancer. We also review potential therapeutic approaches to prevent the development and activation of cancer-associated fibroblasts via inhibition of CAF inducers and CAF markers in cancer.
Collapse
Affiliation(s)
- Ece Ozmen
- Koç University Graduate School of Health Sciences, Istanbul, Türkiye
| | - Tevriz Dilan Demir
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, Türkiye
| |
Collapse
|
5
|
Wang J, Chen C, Huang J, Xie Z, Chen X, Zheng Z, Li E, Zou H. The possibilities of LOXL4 as a prognostic marker for carcinomas. Amino Acids 2023; 55:1519-1529. [PMID: 37814029 DOI: 10.1007/s00726-023-03343-9] [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: 05/16/2022] [Accepted: 09/25/2023] [Indexed: 10/11/2023]
Abstract
Lysyl oxidase-like 4 (LOXL4), a member of lysyl oxidase family, is a copper and lysine tyrosylquinone-dependent amine oxidase that serves the role of catalyzing the cross-linking of elastin and collagen in the extracellular matrix. Numerous studies have shown a significant association between LOXL4 expression levels and tumor proliferation, migration, invasion and patients' prognosis and overall survival in different types of tumors. Here we review their relationship and the molecular pathogenesis behind them, aiming to explore the possibilities of LOXL4 as a prognostic marker for diverse carcinomas and provide some indications for further research in this field.
Collapse
Affiliation(s)
- Jiaming Wang
- Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Chaojian Chen
- Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Jiayi Huang
- Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Ziman Xie
- Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Xiaoxue Chen
- Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Ziqi Zheng
- Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Enmin Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Haiying Zou
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Al-U’datt DGF, Tranchant CC, Al-Husein B, Hiram R, Al-Dwairi A, AlQudah M, Al-shboul O, Jaradat S, Alqbelat J, Almajwal A. Involvement and possible role of transglutaminases 1 and 2 in mediating fibrotic signalling, collagen cross-linking and cell proliferation in neonatal rat ventricular fibroblasts. PLoS One 2023; 18:e0281320. [PMID: 36848364 PMCID: PMC9970086 DOI: 10.1371/journal.pone.0281320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/19/2023] [Indexed: 03/01/2023] Open
Abstract
Transglutaminase (TG) isoforms control diverse normal and pathophysiologic processes through their capacity to cross-link extracellular matrix (ECM) proteins. Their functional and signalling roles in cardiac fibrosis remain poorly understood, despite some evidence of TG2 involvement in abnormal ECM remodelling in heart diseases. In this study, we investigated the role of TG1 and TG2 in mediating fibrotic signalling, collagen cross-linking, and cell proliferation in healthy fibroblasts by siRNA-mediated knockdown. siRNA for TG1, TG2 or negative control was transfected into cultured neonatal rat ventricular fibroblasts and cardiomyocytes. mRNA expression of TGs and profibrotic, proliferation and apoptotic markers was assessed by qPCR. Cell proliferation and soluble and insoluble collagen were determined by ELISA and LC-MS/MS, respectively. TG1 and TG2 were both expressed in neonatal rat cardiomyocytes and fibroblasts before transfection. Other TGs were not detected before and after transfection. TG2 was predominantly expressed and more effectively silenced than TG1. Knocking down TG1 or TG2 significantly modified profibrotic markers mRNA expression in fibroblasts, decreasing connective tissue growth factor (CTGF) and increasing transforming growth factor-β1 compared to the negative siRNA control. Reduced expression of collagen 3A1 was found upon TG1 knockdown, while TG2 knockdown raised α-smooth muscle actin expression. TG2 knockdown further increased fibroblast proliferation and the expression of proliferation marker cyclin D1. Lower insoluble collagen content and collagen cross-linking were evidenced upon silencing TG1 or TG2. Transcript levels of collagen 1A1, fibronectin 1, matrix metalloproteinase-2, cyclin E2, and BCL-2-associated X protein/B-cell lymphoma 2 ratio were strongly correlated with TG1 mRNA expression, whereas TG2 expression correlated strongly with CTGF mRNA abundance. These findings support a functional and signalling role for TG1 and TG2 from fibroblasts in regulating key processes underlying myocardial ECM homeostasis and dysregulation, suggesting that these isoforms could be potential and promising targets for the development of cardiac fibrosis therapies.
Collapse
Affiliation(s)
- Doa’a G. F. Al-U’datt
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Carole C. Tranchant
- School of Food Science, Nutrition and Family Studies, Faculty of Health Sciences and Community Services, Université de Moncton, New Brunswick, Canada
| | - Belal Al-Husein
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Roddy Hiram
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Ahmed Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad AlQudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
- Physiology Department, Arabian Gulf University, Manama, Bahrain
| | - Othman Al-shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Saied Jaradat
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid, Jordan
| | - Jenan Alqbelat
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Liburkin-Dan T, Toledano S, Neufeld G. Lysyl Oxidase Family Enzymes and Their Role in Tumor Progression. Int J Mol Sci 2022; 23:6249. [PMID: 35682926 PMCID: PMC9181702 DOI: 10.3390/ijms23116249] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 02/06/2023] Open
Abstract
The five genes of the lysyl oxidase family encode enzymes that covalently cross-link components of the extracellular matrix, such as various types of collagen and elastin, and, thus, promote the stabilization of extracellular matrixes. Several of these genes, in particular lysyl oxidase (LOX) and lysyl oxidase like-2 (LOXL2) were identified as genes that are upregulated by hypoxia, and promote tumor cells invasion and metastasis. Here, we focus on the description of the diverse molecular mechanisms by which the various lysyl oxidases affect tumor progression. We also describe attempts that have been made, and are still on-going, that focus on the development of efficient lysyl oxidase inhibitors for the treatment of various forms of cancer, and of diseases associated with abnormal fibrosis.
Collapse
Affiliation(s)
| | | | - Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 31096, Israel; (T.L.-D.); (S.T.)
| |
Collapse
|
8
|
Ren P, Niu X, Zhao R, Liu J, Ren W, Dai H, Chen J, Yan J, Li B, Shao Y, Bai Y, Han P. Long non-coding RNA AGAP2-AS1 promotes cell proliferation and invasion through regulating miR-193a-3p/LOXL4 axis in laryngeal squamous cell carcinoma. Cell Cycle 2022; 21:697-707. [PMID: 35113007 PMCID: PMC8973330 DOI: 10.1080/15384101.2021.2016197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is an aggressive malignancy with highly mortality rate. Long non-coding RNA (lncRNA) AGAP2-AS1 is an identified oncogene in several types of cancers. However, the role of AGAP2-AS1 in LSCC remains unclear. The expression levels of AGAP2-AS1 in LSCC tissues and cell lines were measured using qRT-PCR. AGAP2-AS1 was knocked down in LSCC cells through transfection with siRNA-AGAP2-AS1. Cell proliferation and invasion were detected using MTT and transwell assays. Dual-luciferase reporter gene assay was performed to confirm the interaction with AGAP2-AS1 and downstream genes. Our results showed that AGAP2-AS1 expression was remarkably increased in human LSCC tissues and cell lines. Knockdown of AGAP2-AS1 significantly inhibited the proliferation and invasion of LSCC cells. In addition, AGAP2-AS1 sponged miR-193a-3p and regulated its expression in LSCC cells. Inhibition of miR-193a-3p reversed the effects of AGAP2-AS1 knockdown on LSCC cells. Furthermore, Lysyl oxidase-like 4 (LOXL4) was a target gene of miR-193a-3p and the role of miR-193a-3p was mediated by LOXL4. In conclusion, these findings suggest that knockdown of AGAP2-AS1 inhibited the proliferation and invasion of LSCC cells through regulating the miR-193a-3p/LOXL4 axis.
Collapse
Affiliation(s)
- Pengyu Ren
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China,Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Xiaorong Niu
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Ruimin Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Junsong Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Wanli Ren
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Hao Dai
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Jiayu Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Jinfeng Yan
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Baiya Li
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Yuan Shao
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Yanxia Bai
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China,CONTACT Yanxia Bai
| | - Peng Han
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China,Peng Han Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi710061, China
| |
Collapse
|
9
|
Diffuse gastric cancer: Emerging mechanisms of tumor initiation and progression. Biochim Biophys Acta Rev Cancer 2022; 1877:188719. [PMID: 35307354 DOI: 10.1016/j.bbcan.2022.188719] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
Gastric cancer is globally the fourth leading cause of cancer-related deaths. Patients with diffuse-type gastric cancer (DGC) particularly have a poor prognosis that only marginally improved over the last decades, as conventional chemotherapies are frequently ineffective and specific therapies are unavailable. Early-stage DGC is characterized by intramucosal lesions of discohesive cells, which can be present for many years before the emergence of advanced DGC consisting of highly proliferative and invasive cells. The mechanisms underlying the key steps of DGC development and transition to aggressive tumors are starting to emerge. Novel mouse- and organoid models for DGC, together with multi-omic analyses of DGC tumors, revealed contributions of both tumor cell-intrinsic alterations and gradual changes in the tumor microenvironment to DGC progression. In this review, we will discuss how these recent findings are leading towards an understanding of the cellular and molecular mechanisms responsible for DGC initiation and malignancy, which may provide opportunities for targeted therapies.
Collapse
|
10
|
Ji Y, You Y, Wu Y, Wang M, He Q, Zhou X, Chen L, Sun X, Liu Y, Fu X, Kwan HY, Zuo Q, Luo R, Zhao X. Overexpression of miR-328-5p influences cell growth and migration to promote NSCLC progression by targeting LOXL4. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:301. [PMID: 35433959 PMCID: PMC9011230 DOI: 10.21037/atm-22-345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/21/2022] [Indexed: 12/17/2022]
Abstract
Background Lung cancer is the leading cause of cancer-associated mortality worldwide, and most lung cancers are classified as non-small cell lung cancer (NSCLC). MiR-328 influence the progression of multiple tumors, but the role of miR-328-5p in NSCLC has not been elucidated. The aim of this study was to illuminate the oncogenic role and potential molecular mechanisms of the miR-328-5p and lysyl oxidase like 4 (LOXL4) in NSCLC. Methods Expression of miR-328-5p was detected by real-time quantitative polymerase chain reaction (qRT-PCR) in tumor and non-tumor adjacent tissues. After Lentivirus-miR-328-5p was employed to intervene this miRNA in NSCLC cell lines, RT-qPCR was used to detect the expression levels of miR-328-5p. Cell Counting Kit-8 (CCK-8), cell colony formation, flow cytometry, wound healing, Transwell assays were used to determine the malignant phenotypes of NSCLC cells. Nude mice models of subcutaneous tumors were established to observe the effect of miR-328-5p on tumorigenesis. Targeting the 3'UTR of LOXL4 by miR-328-5p was verified by integrated analysis including transcriptome sequencing, dual-luciferase and western-blot assays. Results High miR-328-5p level was observed in NSCLC cells from The Cancer Genome Atlas (TCGA) database and tumor tissues collected from NSCLC patients. Overexpressed miR-328-5p promoted NSCLC cell proliferation, survival, and migration, and promoted tumor growth in vivo. Knockdown of miR-328-5p suppressed tumorigenic activities. Transcriptome sequencing analysis revealed that LOXL4 was downregulated by miR-328-5p, which was confirmed by dual-luciferase reporter and western-blot assays. Conclusions miR-328-5p showed targeted regulation of LOXL4 to promote cell proliferation and migration in NSCLC.
Collapse
Affiliation(s)
- Yanzhao Ji
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Taiyuan, China
| | - Yanting You
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yifen Wu
- Department of Oncology, Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan, China
| | - Min Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Qiuxing He
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xinghong Zhou
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Liqian Chen
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaomin Sun
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yanyan Liu
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiuqiong Fu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Qiang Zuo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ren Luo
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoshan Zhao
- Syndrome Laboratory of Integrated of Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Wang L, Cao S, Zhai R, Zhao Y, Song G. Systematic Analysis of Expression and Prognostic Values of Lysyl Oxidase Family in Gastric Cancer. Front Genet 2022; 12:760534. [PMID: 35126449 PMCID: PMC8812723 DOI: 10.3389/fgene.2021.760534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/16/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Gastric cancer (GC) remains the fifth most commonly diagnosed malignancy worldwide, with a poor prognosis. The lysyl oxidase (LOX) family, a type of secreted copper-dependent amine oxidases, is comprised of LOX and four LOX-like (LOXL) 1–4 isoforms and has been reported to be dysregulated in a number of different type cancers. However, the diverse expression patterns and prognostic values of LOX family in GC have yet to be systematically analyzed. Methods: ONCOMINE, GEPIA, UALCAN, Kaplan–Meier Plotter, LOGpc, cBioPortal, GeneMANIA and Metascape databases were utilized in this study to analyze the expression, prognostic values, mutations and functional networks of LOX family in GC. Results: The mRNA expression levels of LOX, LOXL1 and LOXL2 were significantly higher in GC, the expression level of LOXL3 was contrary in different databases, while the expression level of LOXL4 made no difference; the expression levels of LOX, LOXL1 and LOXL3 were higher in stages 2–4 than that of normal tissues and stage 1, while the mRNA level of LOXL2 in stage 1–4 was higher than normal tissues; patients with high expression of LOX and LOXL 2-4 had poor OS; the genes correlated with LOX and LOXL2 were enriched in extracellular matrix organization, vasculature development and skeletal system development. Conclusion: Our results indicated that the LOX family, especially LOX and LOXL2, might play an important role in GC oncogenesis, and they may become biomarkers for predicting tumor prognosis and potential targets for tumor therapy.
Collapse
Affiliation(s)
- Li Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shan Cao
- Department of Respiratory, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Rujun Zhai
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yang Zhao
- Radiology Department, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guodong Song
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Wang Y, Guo D, Li B, Wang Y, Wang B, Wang Z, Wang M, Teng Q. MiR-665 suppresses the progression of diffuse large B cell lymphoma (DLBCL) through targeting LIM and SH3 protein 1 (LASP1). Leuk Res 2022; 112:106769. [PMID: 34875555 DOI: 10.1016/j.leukres.2021.106769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/05/2021] [Accepted: 11/28/2021] [Indexed: 10/19/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL), the most common type of non-Hodgkin lymphoma worldwide, is aggressive and highly heterogeneous. MiR-665 was found to be lowly expressed in serum exosomes of DLBCL patients and in DLBCL cell lines, but its function in DLBCL progression remains unclear. In this study, miR-665 was overexpressed in SU-DHL-4 cells via miR-665 mimics and knocked down in FARAGE cells via miR-665 inhibitor. Knockdown of miR-665 promoted DLBCL cell proliferation and invasion and decreased cell apoptosis, whereas miR-665 overexpression showed opposite effects on DLBCL cell malignant behaviors. Luciferase reporter assay confirmed LIM and SH3 protein 1 (LASP1) and MYC as target genes of miR-665. Moreover, the expression of LASP1 was negatively correlated with that of miR-665 in LDLBCL cells. LASP1 has tumor-promoting property and its inhibition abolished the effect of miR-665 knockdown on DLBCL cell proliferation, invasion, and apoptosis. SU-DHL-4 cells were inoculated into the flank of nude mice, followed by orthotopic injection with miR-665 agomir. MiR-665 overexpression restricted tumor growth in vivo. Thus, we demonstrates that miR-665 suppresses DLBCL cell malignant behaviors by inhibiting cell proliferation and invasion and inducing apoptosis via targeting LASP1 and MYC, suggesting the importance of miR-665 in DLBCL progression.
Collapse
MESH Headings
- 3' Untranslated Regions/genetics
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Apoptosis/genetics
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cytoskeletal Proteins/genetics
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- HEK293 Cells
- Humans
- LIM Domain Proteins/genetics
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/blood
- MicroRNAs/genetics
- Middle Aged
- Proto-Oncogene Proteins c-myc/genetics
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Yan Wang
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Dongmei Guo
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Banban Li
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Yanyan Wang
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Bo Wang
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Zan Wang
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Meng Wang
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China
| | - Qingliang Teng
- Department of Hematology, Taian City Central Hospital, Taian, Shandong, PR China.
| |
Collapse
|
13
|
In silico identification of the prognostic biomarkers and therapeutic targets associated with cancer stem cell characteristics of glioma. Biosci Rep 2021; 40:225916. [PMID: 32725165 PMCID: PMC7418212 DOI: 10.1042/bsr20201037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/17/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Glioma is the common histological subtype of malignancy in central nervous system, with a high morbidity and mortality. Cancer stem cells (CSCs) play an important role in regulating the tumorigenesis and progression of glioma; however, the prognostic biomarkers and therapeutic targets associated with CSC characteristics have not been fully acknowledged in glioma. In order to identify the prognostic stemness-related genes (SRGs) of glioma in silico, the RNA sequencing data of patients with glioma were retrieved from The Cancer Genome Atlas (TCGA) databases. The mRNA expression-based stemness index (mRNAsi) was significantly associated with the glioma histologic grade, isocitrate dehydrogenase 1 (IDH1) mutation and overall survival of glioma patients by the nonparametric test and Kaplan–Meier survival analysis. A total of 340 SRGs were identified as the overlapped stemness-related differential expressed genes (DEGs) of different histologic grade screened by the univariate Cox analysis. Based on 11 prognostic SRGs, the predict nomogram was constructed with the AUC of 0.832. Moreover, the risk score of the nomogram was an independent prognostic factor, indicating its significant applicability. Besides other eight reported biomarkers of glioma, we found that F2RL2, CLCNKA and LOXL4 were first identified as prognostic biomarkers for glioma. In conclusion, this bioinformatics study demonstrates the mRNAsi as a reliable index for the IDH1 mutation, histologic grade and OS of glioma patients and provides a well-applied model for predicting the OS for patients with glioma based on prognostic SRGs. Additionally, this in silico study also identifies three novel prognostic biomarkers (F2RL2, CLCNKA and LOXL4) for glioma patients.
Collapse
|
14
|
Targeting Lysyl Oxidase Family Meditated Matrix Cross-Linking as an Anti-Stromal Therapy in Solid Tumours. Cancers (Basel) 2021; 13:cancers13030491. [PMID: 33513979 PMCID: PMC7865543 DOI: 10.3390/cancers13030491] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary To improve efficacy of solid cancer treatment, efforts have shifted towards targeting both the cancer cells and the surrounding tumour tissue they grow in. The lysyl oxidase (LOX) family of enzymes underpin the fibrotic remodeling of the tumour microenvironment to promote both cancer growth, spread throughout the body and modulate response to therapies. This review examines how the lysyl oxidase family is involved in tumour development, how they can be targeted, and their potential as diagnostic and prognostic biomarkers in solid tumours. Abstract The lysyl oxidase (LOX) family of enzymes are a major driver in the biogenesis of desmoplastic matrix at the primary tumour and secondary metastatic sites. With the increasing interest in and development of anti-stromal therapies aimed at improving clinical outcomes of cancer patients, the Lox family has emerged as a potentially powerful clinical target. This review examines how lysyl oxidase family dysregulation in solid cancers contributes to disease progression and poor patient outcomes, as well as an evaluation of the preclinical landscape of LOX family targeting therapeutics. We also discuss the suitability of the LOX family as a diagnostic and/or prognostic marker in solid tumours.
Collapse
|
15
|
Luo C, Hu C, Li B, Liu J, Hu L, Dong R, Liao X, Zhou J, Xu L, Liu S, Li Y, Yuan D, Jiang W, Yan J. Polymorphisms in Lysyl Oxidase Family Genes Are Associated With Intracranial Aneurysm Susceptibility in a Chinese Population. Front Endocrinol (Lausanne) 2021; 12:642698. [PMID: 34393991 PMCID: PMC8355735 DOI: 10.3389/fendo.2021.642698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 06/29/2021] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Intracranial aneurysms (IA) comprise a multifactorial disease with unclear physiological mechanisms. The lysyl oxidase (LOX) family genes (LOX, LOX-like 1-4) plays important roles in extracellular matrix (ECM) reconstruction and has been investigated in terms of susceptibility to IA in a few populations. We aimed to determine whether polymorphisms in LOX family genes are associated with susceptibility to IA in a Chinese population. METHODS This case-control study included 384 patients with IA and 384 healthy individuals without IA (controls). We genotyped 27 single nucleotide polymorphisms (SNPs) of LOX family genes using the Sequenom MassARRAY® platform. These SNPs were adjusted for known risk factors and then, odds ratios (OR) and 95% confidence intervals (CI) were evaluated using binary logistic regression analysis. RESULTS The result showed that LOX rs10519694 was associated with the risk of IA in recessive (OR, 3.88; 95% CI, 1.12-13.47) and additive (OR, 1.56; 95%CI, 1.05-2.34) models. Stratified analyses illustrated that LOX rs10519694 was associated with the risk of single IA in the recessive (OR, 3.95; 95%CI, 1.04-15.11) and additive (OR, 1.64; 95%CI, 1.04-2.56) models. The LOXL2 rs1010156 polymorphism was associated with multiple IA in the dominant model (OR, 1.92; 95%CI, 1.02-3.62). No associations were observed between SNPs of LOXL1, LOXL3, and LOXL4 and risk of IA. CONCLUSION LOX and LOXL2 polymorphisms were associated with risk of single IA and multiple IA in a Chinese population, suggesting potential roles of these genes in IA. The effects of these genes on IA require further investigation.
Collapse
Affiliation(s)
- Chun Luo
- Department of Epidemiology and Health Statistics, XiangYa School of Public Health, Central South University, Changsha, China
| | - Chongyu Hu
- Department of Neurology, Hunan People’s Hospital, Changsha, China
| | - Bingyang Li
- Department of Epidemiology and Health Statistics, XiangYa School of Public Health, Central South University, Changsha, China
- Department of Information Statistics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eight Hospital), Changsha, China
| | - Junyu Liu
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Liming Hu
- Department of Epidemiology and Health Statistics, XiangYa School of Public Health, Central South University, Changsha, China
| | - Rui Dong
- Department of Epidemiology and Health Statistics, XiangYa School of Public Health, Central South University, Changsha, China
| | - Xin Liao
- Department of Epidemiology and Health Statistics, XiangYa School of Public Health, Central South University, Changsha, China
- Department of Scientific Research, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jilin Zhou
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Lu Xu
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Songlin Liu
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Yifeng Li
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Dun Yuan
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Weixi Jiang
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, China
| | - Junxia Yan
- Department of Epidemiology and Health Statistics, XiangYa School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, XiangYa School of Public Health, Central South University, Changsha, China
- *Correspondence: Junxia Yan,
| |
Collapse
|
16
|
Ye M, Zhou J, Gao Y, Pan S, Zhu X. The prognostic value of the lysyl oxidase family in ovarian cancer. J Clin Lab Anal 2020; 34:e23538. [PMID: 33058284 PMCID: PMC7755792 DOI: 10.1002/jcla.23538] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Our study intended to evaluate the prognostic value of lysyl oxidase (LOX) and its four relevant members, the lysyl oxidase-like genes (LOXL1-4), in ovarian cancer (OC) patients. MATERIAL AND METHODS The Kaplan-Meier plotter (KM plotter) database was used to investigate the prognostic power of the LOX family for OC patients. Overall survival (OS) and progression-free survival (PFS) were the clinical endpoints. The prognostic roles of the LOX family in OC patients were also analyzed according to various clinicopathological characteristics, including histological subtypes, clinical stages, pathological grades, and chemotherapeutic treatments. RESULTS Overexpression of LOX, LOXL1, LOXL2, and LOXL3 mRNA indicated poor OS and PFS in OC patients, particularly in serous and grade II + III OC patients. Overexpression of LOXL4 mRNA resulted in worse PFS in OC patients. Overexpression of LOX and LOXL1 mRNA showed worse OS and PFS in stage III + IV OC patients, and overexpression of LOXL3 mRNA indicated worse OS and PFS in stage I + II OC patients. Overexpression of LOX, LOXL3, and LOXL4 mRNA indicated worse OS and PFS among OC patients who received platinum, taxol, and taxol + platinum chemotherapy. Overexpression of LOXL1 and LOXL2 mRNA was related to lower OS and PFS in OC patients who received platinum chemotherapy. CONCLUSION LOX, LOXL1, LOXL2, and LOXL3 may become potential predictive markers for negative outcomes in OC patients. Moreover, the LOX family can serve as new molecular predictors for the efficiency of platinum-based chemotherapy in OC patients.
Collapse
Affiliation(s)
- Miaomiao Ye
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Junhan Zhou
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ying Gao
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shuya Pan
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xueqiong Zhu
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
17
|
Ye M, Song Y, Pan S, Chu M, Wang ZW, Zhu X. Evolving roles of lysyl oxidase family in tumorigenesis and cancer therapy. Pharmacol Ther 2020; 215:107633. [PMID: 32693113 DOI: 10.1016/j.pharmthera.2020.107633] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
The lysyl oxidase (LOX) family is comprised of LOX and four LOX-like proteins (LOXL1, LOXL2, LOXL3, and LOXL4), and mainly functions in the remodeling of extracellular matrix (ECM) and the cross-linking of collagen and elastic fibers. Recently, a growing body of research has demonstrated that LOX family is critically involved in the regulation of cancer cell proliferation, migration, invasion and metastasis. In this review, we discuss the roles of LOX family members in the development and progression of different types of human cancers. Furthermore, we also describe the potential inhibitors of LOX family proteins and highlight that LOX family might be an important therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shuya Pan
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Man Chu
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhi-Wei Wang
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China..
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
18
|
Selmansberger M, Michna A, Braselmann H, Höfig I, Schorpp K, Weber P, Anastasov N, Zitzelsberger H, Hess J, Unger K. Transcriptome network of the papillary thyroid carcinoma radiation marker CLIP2. Radiat Oncol 2020; 15:182. [PMID: 32727620 PMCID: PMC7392692 DOI: 10.1186/s13014-020-01620-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/15/2020] [Indexed: 11/29/2022] Open
Abstract
Background We present a functional gene association network of the CLIP2 gene, generated by de-novo reconstruction from transcriptomic microarray data. CLIP2 was previously identified as a potential marker for radiation induced papillary thyroid carcinoma (PTC) of young patients in the aftermath of the Chernobyl reactor accident. Considering the rising thyroid cancer incidence rates in western societies, potentially related to medical radiation exposure, the functional characterization of CLIP2 is of relevance and contributes to the knowledge about radiation-induced thyroid malignancies. Methods We generated a transcriptomic mRNA expression data set from a CLIP2-perturbed thyroid cancer cell line (TPC-1) with induced CLIP2 mRNA overexpression and siRNA knockdown, respectively, followed by gene-association network reconstruction using the partial correlation-based approach GeneNet. Furthermore, we investigated different approaches for prioritizing differentially expressed genes for network reconstruction and compared the resulting networks with existing functional interaction networks from the Reactome, Biogrid and STRING databases. The derived CLIP2 interaction partners were validated on transcript and protein level. Results The best reconstructed network with regard to selection parameters contained a set of 20 genes in the 1st neighborhood of CLIP2 and suggests involvement of CLIP2 in the biological processes DNA repair/maintenance, chromosomal instability, promotion of proliferation and metastasis. Peptidylprolyl Isomerase Like 3 (PPIL3), previously identified as a potential direct interaction partner of CLIP2, was confirmed in this study by co-expression at the transcript and protein level. Conclusion In our study we present an optimized preselection approach for genes subjected to gene-association network reconstruction, which was applied to CLIP2 perturbation transcriptome data of a thyroid cancer cell culture model. Our data support the potential carcinogenic role of CLIP2 overexpression in radiation-induced PTC and further suggest potential interaction partners of the gene.
Collapse
Affiliation(s)
- Martin Selmansberger
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Agata Michna
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Herbert Braselmann
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Ines Höfig
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Kenji Schorpp
- Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Peter Weber
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Natasa Anastasov
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Horst Zitzelsberger
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany. .,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany. .,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.
| |
Collapse
|
19
|
Yin H, Wang Y, Wu Y, Zhang X, Zhang X, Liu J, Wang T, Fan J, Sun J, Yang A, Zhang R. EZH2-mediated Epigenetic Silencing of miR-29/miR-30 targets LOXL4 and contributes to Tumorigenesis, Metastasis, and Immune Microenvironment Remodeling in Breast Cancer. Theranostics 2020; 10:8494-8512. [PMID: 32754259 PMCID: PMC7392008 DOI: 10.7150/thno.44849] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/21/2020] [Indexed: 12/14/2022] Open
Abstract
Enhancer of Zeste Homolog 2 (EZH2), a key epigenetic regulator, is involved in breast cancer progression and metastasis. LOXL4 is increasingly recognized as an important player in cancer progression. To date, how EZH2 regulates LOXL4 in the progression of breast cancer remains unclear. Methods: We evaluated the association between LOX family proteins and EZH2 in invasive breast carcinoma through the starBase v2.0 analysis, and its correlation with breast tumorigenesis using the Oncomine dataset. We then applied miRcode data combined with gene expression omnibus (GEO) data to screen candidate miRNAs mediating the regulation of LOXL4 by EZH2. We explored the regulatory mechanism of EZH2, miR-29b/miR-30d, and LOXL4 in breast cancer cells by qRT-PCR, Western blotting, cell proliferation, colony formation, and wound healing assays, xenograft experiments, dual-luciferase reporter assay, and chromatin immunoprecipitation. All statistical tests were two-sided. Results: Inhibition of EZH2 or LOXL4, or miR-29b/miR-30d overexpression, decreased breast cancer cell proliferation, migration, and metastasis in vitro and in vivo. LOXL4 was identified as a direct target of miR-29b and miR-30d. EZH2 inhibition enhanced miR-30d and miR-29b transcription via promoter binding activity, leading to the reduced expression of LOXL4. Immunohistochemical analysis of human breast cancer specimens and flow cytometry analysis of tumor-infiltrating macrophages in mice showed a positive association of EZH2 with LOXL4 expression and macrophage infiltration. Conclusions: Our findings identified EZH2-miR-29b/miR-30d-LOXL4 signaling pathway was involved in breast tumorigenesis, and suggested that the epigenetic modulation represents a potential therapeutic target for breast cancer by controlling macrophage activation.
Collapse
|
20
|
Abbaszadegan MR, Mojarrad M, Moghbeli M. Role of extra cellular proteins in gastric cancer progression and metastasis: an update. Genes Environ 2020; 42:18. [PMID: 32467737 PMCID: PMC7227337 DOI: 10.1186/s41021-020-00157-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common cancers in the world with a high ratio of mortality. Regarding the late diagnosis, there is a high ratio of distant metastasis among GC cases. Despite the recent progresses in therapeutic modalities, there is not still an efficient therapeutic method to increase survival rate of metastatic GC cases. Main body Apart from the various intracellular signaling pathways which are involved in tumor cell migration and metastasis, the local microenvironment is also a critical regulator of tumor cell migration. Indeed, the intracellular signaling pathways also exert their final metastatic roles through regulation of extra cellular matrix (ECM). Therefore, it is required to assess the role of extra cellular components in biology of GC. Conclusion In the present review, we summarize 48 of the significant ECM components including 17 ECM modifying enzymes, seven extracellular angiogenic factors, 13 cell adhesion and cytoskeletal organizers, seven matricellular proteins and growth factors, and four proteoglycans and extra cellular glycoproteins. This review paves the way of determination of a specific extra cellular diagnostic and prognostic panel marker for the GC patients.
Collapse
Affiliation(s)
| | - Majid Mojarrad
- 2Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- 2Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Yuzhalin AE, Lim SY, Gordon-Weeks AN, Fischer R, Kessler BM, Yu D, Muschel RJ. Proteomics analysis of the matrisome from MC38 experimental mouse liver metastases. Am J Physiol Gastrointest Liver Physiol 2019; 317:G625-G639. [PMID: 31545917 PMCID: PMC6879896 DOI: 10.1152/ajpgi.00014.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 01/31/2023]
Abstract
Dissemination of primary tumors to distant anatomical sites has a substantial negative impact on patient prognosis. The liver is a common site for metastases from colorectal cancer, and patients with hepatic metastases have generally much shorter survival, raising a need to develop and implement novel strategies for targeting metastatic disease. The extracellular matrix (ECM) is a meshwork of highly crosslinked, insoluble high-molecular-mass proteins maintaining tissue integrity and establishing cell-cell interactions. Emerging evidence identifies the importance of the ECM in cancer cell migration, invasion, intravasation, and metastasis. Here, we isolated the ECM from MC38 mouse liver metastases using our optimized method of mild detergent solubilization followed by biochemical enrichment. The matrices were subjected to label-free quantitative mass spectrometry analysis, revealing proteins highly abundant in the metastatic matrisome. The resulting list of proteins upregulated in the ECM significantly predicted survival in patients with colorectal cancer but not other cancers with strong involvement of the ECM component. One of the proteins upregulated in liver metastatic ECM, annexin A1, was not previously studied in the context of cancer-associated matrisome. Here, we show that annexin A1 was markedly upregulated in colon cancer cell lines compared with cancer cells of other origin and also over-represented in human primary colorectal lesions, as well as hepatic metastases, compared with their adjacent healthy tissue counterparts. In conclusion, our study provides a comprehensive ECM characterization of MC38 experimental liver metastases and proposes annexin A1 as a putative target for this disease.NEW & NOTEWORTHY Here, the authors provide an extensive proteomics characterization of murine colorectal cancer liver metastasis matrisome (the ensemble of all extracellular matrix molecules). The findings presented in this study may enable identification of therapeutic targets or biomarkers of hepatic metastases.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Cancer Research United Kingdom/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Su Yin Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Alex N Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ruth J Muschel
- Cancer Research United Kingdom/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Vélez JI, Lopera F, Silva CT, Villegas A, Espinosa LG, Vidal OM, Mastronardi CA, Arcos-Burgos M. Familial Alzheimer's Disease and Recessive Modifiers. Mol Neurobiol 2019; 57:1035-1043. [PMID: 31664702 PMCID: PMC7031188 DOI: 10.1007/s12035-019-01798-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/22/2019] [Indexed: 12/15/2022]
Abstract
Alzheimer’s disease (AD) is progressive brain disorder that affects ~ 50 million people worldwide and has no current effective treatment. AD age of onset (ADAOO) has shown to be critical for the identification of genes that modify the appearance of AD signs and symptoms in a specific population. We clinically characterized and whole-exome genotyped 71 individuals with AD from the Paisa genetic isolate, segregating the (PSEN1) E280A dominant fully penetrant mutation, and analyzed the potential recessive effects of ~ 50,000 common functional genomic variants to the ADAOO. Standard quality control and filtering procedures were applied, and recessive single- and multi-locus linear mixed-effects models were used. We identified genetic variants in the SLC9C1, CSN1S1, and LOXL4 acting recessively to delay ADAOO up to ~ 11, ~ 6, and ~ 9 years on average, respectively. In contrast, the CC recessive genotype in marker DHRS4L2-rs2273946 accelerates ADAOO by ~ 8 years. This study, reports new recessive variants modifying ADAOO in PSEN1 E280A mutation carriers. This set of genes are implicated in important biological processes and molecular functions commonly affected by genes associated with the etiology of AD such as APP, APOE, and CLU. Future functional studies using modern techniques such as induced pluripotent stem cells will allow a better understanding of the over expression and down regulation of these recessive modifier variants and hence the pathogenesis of AD. These results are important for prediction of AD and ultimately, substantial to develop new therapeutic strategies for individuals at risk or affected by AD.
Collapse
Affiliation(s)
| | - Francisco Lopera
- Neuroscience Research Group, University of Antioquia, Medellín, Colombia
| | - Claudia T Silva
- Neuroscience Research Group, University of Antioquia, Medellín, Colombia
| | - Andrés Villegas
- Neuroscience Research Group, University of Antioquia, Medellín, Colombia
| | - Lady G Espinosa
- INPAC Research Group, Fundación Universitaria Sanitas, Bogotá, Colombia
| | | | | | - Mauricio Arcos-Burgos
- Grupo de Investigación en Psiquiatría (GIPSI), Departamento de Psiquiatría, Instituto de Investigaciones Médicas (IIM), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
23
|
Lysyl oxidases: linking structures and immunity in the tumor microenvironment. Cancer Immunol Immunother 2019; 69:223-235. [PMID: 31650200 PMCID: PMC7000489 DOI: 10.1007/s00262-019-02404-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/24/2019] [Indexed: 02/08/2023]
Abstract
The lysyl oxidases (LOXs) are a family of enzymes deputed to cross-link collagen and elastin, shaping the structure and strength of the extracellular matrix (ECM). However, many novel “non-canonical” functions, alternative substrates, and regulatory mechanisms have been described and are being continuously elucidated. The activity of LOXs, therefore, appears to be integrated into a complex network of signals regulating many cell functions, including survival/proliferation/differentiation. Among these signaling pathways, TGF-β and PI3K/Akt/mTOR, in particular, cross-talk extensively with each other and with LOXs also initiating complex feedback loops which modulate the activity of LOXs and direct the remodeling of the ECM. A growing body of evidence indicates that LOXs are not only important in the homeostasis of the normal structure of the ECM, but are also implicated in the establishment and maturation of the tumor microenvironment. LOXs’ association with advanced and metastatic cancer is well established; however, there is enough evidence to support a significant role of LOXs in the transformation of normal epithelial cells, in the accelerated tumor development and the induction of invasion of the premalignant epithelium. A better understanding of LOXs and their interactions with the different elements of the tumor immune microenvironment will prove invaluable in the design of novel anti-tumor strategies.
Collapse
|
24
|
Tang H, Long Q, Zhuang K, Yan Y, Han K, Guo H, Lu X. miR-665 promotes the progression of gastric adenocarcinoma via elevating FAK activation through targeting SOCS3 and is negatively regulated by lncRNA MEG3. J Cell Physiol 2019; 235:4709-4719. [PMID: 31650535 DOI: 10.1002/jcp.29349] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
Abstract
Studies have found that miR-665 acted as a tumor suppressor or an oncogene in different malignancies. miR-665 expression was elevated in gastric adenocarcinoma tissues; however, its role and mechanism in this disease are not fully clarified. The expression of miR-665 and its target gene was detected in human gastric adenocarcinoma tissues and cells. Moreover, we analyzed the effects of miR-665 on the proliferation, migration, and epithelial-mesenchymal transition (EMT) of gastric adenocarcinoma cells as well as tumor growth in vivo. The mechanisms of miR-665 in gastric adenocarcinoma were investigated by using molecular biology techniques. We found miR-665 was upregulated and suppressor of cytokine signaling 3 (SOCS3) was downregulated in gastric adenocarcinoma tissues and cells. Elevated miR-665 was positively correlated with tumor size, lymph node metastasis, invasion depth, TNM stage, and poor differentiation in gastric adenocarcinoma patients. Overexpression of miR-665 promoted, whereas knockdown of miR-665 suppressed the proliferation, migration, and EMT of gastric adenocarcinoma cells. Furthermore, we demonstrated that miR-665 functioned through targeting SOCS3, followed by activation of the FAK/Src signaling pathway in gastric adenocarcinoma cells. miR-665 antagomir inhibited tumor growth as well as the activation of the FAK/Src pathway but increased SOCS3 expression in nude mice. In addition, miR-665 expression was negatively regulated by long noncoding RNA maternally expressed gene 3 (MEG3). In conclusion, miR-665 acted as an oncogene in gastric adenocarcinoma by inhibiting SOCS3 followed by activation of the FAK/Src pathway and it was negatively modulated by MEG3. miR-665 may be a promising therapeutic target for the treatment of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Hailing Tang
- Division of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Qianfa Long
- Division of Neurosurgery, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Kun Zhuang
- Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Yuan Yan
- Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Kun Han
- Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Hanqing Guo
- Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Xiaolan Lu
- Division of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
25
|
Yeo MS, Subhash VV, Suda K, Balcıoğlu HE, Zhou S, Thuya WL, Loh XY, Jammula S, Peethala PC, Tan SH, Xie C, Wong FY, Ladoux B, Ito Y, Yang H, Goh BC, Wang L, Yong WP. FBXW5 Promotes Tumorigenesis and Metastasis in Gastric Cancer via Activation of the FAK-Src Signaling Pathway. Cancers (Basel) 2019; 11:cancers11060836. [PMID: 31213005 PMCID: PMC6627937 DOI: 10.3390/cancers11060836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/26/2022] Open
Abstract
F-box/WD repeat-containing protein 5 (FBXW5) is a member of the FBXW subclass of F-box proteins. Despite its known function as a component of the Skp1-Cullin-F-box (SCF) ubiquitin ligase complex, the role of FBXW5 in gastric cancer tumorigenesis and metastasis has not been investigated. The present study investigates the role of FBXW5 in tumorigenesis and metastasis, as well as the regulation of key signaling pathways in gastric cancer; using in-vitro FBXW5 knockdown/overexpression cell line and in-vivo models. In-vitro knockdown of FBXW5 results in a decrease in cell proliferation and cell cycle progression, with a concomitant increase in cell apoptosis and caspase-3 activity. Furthermore, knockdown of FBXW5 also leads to a down regulation in cell migration and adhesion, characterized by a reduction in actin polymerization, focal adhesion turnover and traction forces. This study also delineates the mechanistic role of FBXW5 in oncogenic signaling as its inhibition down regulates RhoA-ROCK 1 (Rho-associated protein kinase 1) and focal adhesion kinase (FAK) signaling cascades. Overexpression of FBXW5 promotes in-vivo tumor growth, whereas its inhibition down regulates in-vivo tumor metastasis. When considered together, our study identifies the novel oncogenic role of FBXW5 in gastric cancer and draws further interest regarding its clinical utility as a potential therapeutic target.
Collapse
Affiliation(s)
- Mei Shi Yeo
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore 119228, Singapore.
| | - Vinod Vijay Subhash
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
- Lowy Cancer Research Centre, University of New South Wales, Sydney 20152, Australia.
| | - Kazuto Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Hayri Emrah Balcıoğlu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.
| | - Siqin Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Win Lwin Thuya
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Xin Yi Loh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Sriganesh Jammula
- Cancer Research UK Cambridge Institute, University of Cambridge, CB2 0RE Cambridge, UK.
| | - Praveen C Peethala
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Shi Hui Tan
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore 119228, Singapore.
| | - Chen Xie
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore 119228, Singapore.
| | - Foong Ying Wong
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore 119228, Singapore.
| | - Benoit Ladoux
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.
- Institut Jacques Monod, Centre National de la Recherche Scientifique, CNRS UMR 7592, Université Paris-Diderot, CEDEX 13, 75205 Paris, France.
| | - Yoshiaki Ito
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore 119228, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore 119228, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.
| |
Collapse
|
26
|
Amendola PG, Reuten R, Erler JT. Interplay Between LOX Enzymes and Integrins in the Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11050729. [PMID: 31130685 PMCID: PMC6562985 DOI: 10.3390/cancers11050729] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/24/2022] Open
Abstract
Members of the lysyl oxidase (LOX) family are secreted copper-dependent amine oxidases that catalyze the covalent crosslinking of collagens and elastin in the extracellular matrix (ECM), an essential process for the structural integrity of all tissues. LOX enzymes can also remodel the tumor microenvironment and have been implicated in all stages of tumor initiation and progression of many cancer types. Changes in the ECM can influence several cancer cell phenotypes. Integrin adhesion complexes (IACs) physically connect cells with their microenvironment. This review article summarizes the main findings on the role of LOX proteins in modulating the tumor microenvironment, with a particular focus on how ECM changes are integrated by IACs to modulate cells behavior. Finally, we discuss how the development of selective LOX inhibitors may lead to novel and effective therapies in cancer treatment.
Collapse
Affiliation(s)
- Pier Giorgio Amendola
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Raphael Reuten
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Janine Terra Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
27
|
Xie W, Huang P, Wu B, Chen S, Huang Z, Wang J, Sun H, Wu J, Xie L, Cheng Y, Xie W, Xu L, Chen LQ, Li E, Zou H. Clinical significance of LOXL4 expression and features of LOXL4-associated protein-protein interaction network in esophageal squamous cell carcinoma. Amino Acids 2019; 51:813-828. [PMID: 30900087 DOI: 10.1007/s00726-019-02723-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/12/2019] [Indexed: 02/05/2023]
Abstract
Lysyl oxidase-like 4 (LOXL4), a member of the LOX family proteins, catalyzes oxidative deamination of lysine residues in collagen and elastin, which are responsible for maintaining extracellular matrix homeostasis. In this study, the mRNA expression of LOXL4 in seven esophageal squamous cell carcinoma (ESCC) cell lines and 15 ESCC pairs of clinical samples were examined. Furthermore, LOXL4 protein levels in the ESCC cell lines were determined using western blotting. With the use of immunofluorescence, LOXL4 was observed to be localized primarily in the cytoplasm, but was also present in the nucleus. In addition, the results indicated that the upregulated expression of LOXL4 was associated with poor survival in patients with ESCC even following curative resection (P = 0.010). Similar Kaplan-Meier estimator curves for proteins that interact with LOXL4, SUV39H1 (P = 0.014) and COL2A1 (P = 0.011), were plotted. The analyses based on the protein-protein interaction network depicted the expression of LOXL4 and its associated proteins as well as their functions, suggesting that LOXL4 and its associated proteins may serve a significant role in the development and progression of ESCC. In conclusion, the results of the present study suggest that LOXL4 is a potential biomarker for patients with ESCC, as well as SUV39H1 and COL2A1, and high expression levels of these genes are associated with poor prognosis in patients with ESCC.
Collapse
Affiliation(s)
- Weijie Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Peiqi Huang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Bingli Wu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Sijie Chen
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Zijian Huang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Junhao Wang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Hong Sun
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Jianyi Wu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Lei Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Yinwei Cheng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Wenming Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Medical Bioinformatics Center, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Liyan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Long-Qi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Enmin Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China.
| | - Haiying Zou
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
28
|
Hong X, Yu JJ. Silencing of lysyl oxidase‑like 2 inhibits the migration, invasion and epithelial‑to‑mesenchymal transition of renal cell carcinoma cells through the Src/FAK signaling pathway. Int J Oncol 2019; 54:1676-1690. [PMID: 30816490 PMCID: PMC6438419 DOI: 10.3892/ijo.2019.4726] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to investigate the effects of lysyl oxidase-like 2 (LOXL2) on the invasion, migration and epithelial-to-mesenchymal transition (EMT) of renal cell carcinoma (RCC) cells through the steroid receptor coactivator (Src)/focal adhesion kinase (FAK) signaling pathway. RCC tissues and adjacent normal tissues were collected from 80 patients with RCC. Immunohistochemistry was used to determine the positive expression rate of the LOXL2 protein. The expression levels of LOXL2 in the HK-2, 786-O, ACHN, Caki1 and A498 cell lines were detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The high LOXL2-expressing 786-O cells were selected for gene silencing experiments, whereas Caki1 cells, which exhibited low LOXL2 expression, were used for overexpression experiments. RT-qPCR and western blot analysis were applied to determine the expression of LOXL2, FAK, Src, matrix metalloproteinase (MMP)-9, epithelial (E)-cadherin, neuronal (N)-cadherin and vimentin. A MTT assay, a Transwell assay, a wound healing assay and flow cytometry were performed to detect cell proliferation, invasion, migration, cell cycle distribution and apoptosis, respectively. The protein expression rate of LOXL2 in RCC tissues was higher compared with that in adjacent normal tissues. Compared with adjacent normal tissues, the mRNA and protein expression levels of LOXL2, FAK, Src, MMP-9, N-cadherin and vimentin and the levels of FAK and Src phosphorylation were increased, while the mRNA and protein expression levels of E-cadherin were decreased in RCC tissues. Following the transfection of 786-O cells with small interfering (si) RNA against LOXL2, the mRNA and protein expression levels of FAK, Src, MMP-9, N-cadherin and vimentin and the levels of phosphorylated FAK and Src were notably decreased in the si-LOXL2 and PP2 inhibitor treated groups, while that of E-cadherin was substantially increased. Additionally, cell proliferation, invasion, migration and the percentage of RCC cells in the G1 phase were reduced, and cell apoptosis was increased. Additionally, Caki1 cells transfected with LOXL2 exhibited an opposite trend. In summary, these results indicate that LOXL2 silencing inhibits the invasion, migration and EMT in RCC cells through inhibition of the Src/FAK signaling pathway.
Collapse
Affiliation(s)
- Xi Hong
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jian-Jun Yu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
29
|
Qu Y, Dou P, Hu M, Xu J, Xia W, Sun H. circRNA‑CER mediates malignant progression of breast cancer through targeting the miR‑136/MMP13 axis. Mol Med Rep 2019; 19:3314-3320. [PMID: 30816475 DOI: 10.3892/mmr.2019.9965] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/13/2019] [Indexed: 11/05/2022] Open
Abstract
Chondrocyte extracellular matrix‑related circular RNAs (circRNA‑CER) have been demonstrated to be involved in various diseases. However, its role in the development of human breast cancer is not clearly understood. The aims of the present study were to assess circRNA‑CER expression in paired cancer tissue and adjacent non‑tumor tissue from 24 patients with breast cancer, and to explore the roles and mechanisms by which circRNA‑CER mediates the malignant progression of breast cancer cells. The results revealed that circRNA‑CER and matrix metalloproteinase 13 (MMP13) were upregulated, whereas miR‑136 was downregulated in breast cancer tissues compared with adjacent non‑tumor tissues. In vitro silencing of circRNA‑CER using small interfering RNA (siRNA) had inhibitory effects on MCF‑7 breast cancer cell proliferation and migration, and similar results were obtained following overexpression of microRNA (miR)‑136 in MCF‑7 cells by transfection with miR‑136 mimics. The subsequent mechanistic study revealed that the expression levels of MMP13 were significantly lower in MCF‑7 cells following transfection with miR‑136 mimics, and silencing of circRNA‑CER enhanced miR‑136 and decreased MMP13 expression levels. Furthermore, silencing of miR‑136 by transfection with miR‑136 inhibitors resulted in an increase in MCF‑7 cell proliferation and migration. miR‑136 inhibitor‑derived biological effects were reversed by co‑transfection of cells with miR‑136 inhibitors and circRNA‑CER siRNA. Taken together, the present results suggested that circRNA‑CER may serve an important role in the progression of breast cancer by regulating the activity of the miR‑136/MMP13 axis, and may be a potential biomarker for the prediction and treatment of breast cancer.
Collapse
Affiliation(s)
- Yikun Qu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Penghui Dou
- Department of Radiation and Chemotherapy, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Ming Hu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Jian Xu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Weibin Xia
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Hongmei Sun
- Department of Oncology, Oncology Hospital of Jiamusi City, Jiamusi, Heilongjiang 154002, P.R. China
| |
Collapse
|
30
|
Shao J, Lu J, Zhu W, Yu H, Jing X, Wang YL, Wang X, Wang XJ. Derepression of LOXL4 inhibits liver cancer growth by reactivating compromised p53. Cell Death Differ 2019; 26:2237-2252. [PMID: 30728460 PMCID: PMC6889417 DOI: 10.1038/s41418-019-0293-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/21/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023] Open
Abstract
TP53 is the most frequently mutated gene in human cancer, whereas tumors with wild-type TP53 develop alternative strategies to survive. Identifying new regulators of p53 reactivation would greatly contribute to the development of cancer therapies. After screening the entire genome in liver cancer cells, we identified lysyl oxidase-like 4 (LOXL4) as a novel regulator for p53 activation. We found that 5-azacytidine (5-aza-CR) induces LOXL4 upregulation, with LOXL4 subsequently binding the basic domain of p53 via its low-isoelectric point region. The interaction between LOXL4 and p53 induces the reactivation of compromised p53, resulting in cell death. Furthermore, the nude mouse xenograft model showed that the 5-aza-CR-dependent LOXL4-p53 axis reduces tumor growth. A positive correlation between LOXL4 expression and overall survival in liver cancer patients with wild-type p53 tumors was observed. In conclusion, we found that 5-aza-CR-induced LOXL4 upregulation reactivates wild-type p53 and triggers cell death, which blocks liver cancer development.
Collapse
Affiliation(s)
- Jialiang Shao
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China.,Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Jiongjiong Lu
- Department of Special Treatment and Liver Transplantation, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Wencheng Zhu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Hua Yu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Xiaoqian Jing
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi-Lin Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China.
| | - Xiong-Jun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China. .,Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China.
| |
Collapse
|
31
|
Li R, Wang Y, Zhang X, Feng M, Ma J, Li J, Yang X, Fang F, Xia Q, Zhang Z, Shang M, Jiang S. Exosome-mediated secretion of LOXL4 promotes hepatocellular carcinoma cell invasion and metastasis. Mol Cancer 2019; 18:18. [PMID: 30704479 PMCID: PMC6354392 DOI: 10.1186/s12943-019-0948-8] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/24/2019] [Indexed: 12/12/2022] Open
Abstract
Background Lysyl oxidase-like 4 (LOXL4) has been found to be dysregulated in several human malignancies, including hepatocellular carcinoma (HCC). However, the role of LOXL4 in HCC progression remains largely unclear. In this study, we investigated the clinical significance and biological involvement of LOXL4 in the progression of HCC. Methods LOXL4 expression was measured in HCC tissues and cell lines. Overexpression, shRNA-mediated knockdown, recombinant human LOXL4 (rhLOXL4), and deletion mutants were applied to study the function of LOXL4 in HCC. Exosomes derived from HCC cell lines were assessed for the ability to promote cancer progression in standard assays. The effects of LOXL4 on the FAK/Src pathway were examined by western blotting. Results LOXL4 was commonly upregulated in HCC tissues and predicted a poor prognosis. Elevated LOXL4 was associated with tumor differentiation, vascular invasion, and tumor-node-metastasis (TNM) stage. Overexpression of LOXL4 promoted, whereas knockdown of LOXL4 inhibited cell migration and invasion of HCC in vitro, and overexpressed LOXL4 promoted intrahepatic and pulmonary metastases of HCC in vivo. Most interestingly, we found that HCC-derived exosomes transferred LOXL4 between HCC cells, and intracellular but not extracellular LOXL4 promoted cell migration by activating the FAK/Src pathway dependent on its amine oxidase activity through a hydrogen peroxide-mediated mechanism. In addition, HCC-derived exosomes transferred LOXL4 to human umbilical vein endothelial cells (HUVECs) though a paracrine mechanism to promote angiogenesis. Conclusions Taken together, our data demonstrate a novel function of LOXL4 in tumor metastasis mediated by exosomes through regulation of the FAK/Src pathway and angiogenesis in HCC. Electronic supplementary material The online version of this article (10.1186/s12943-019-0948-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rongkun Li
- Department of Interventional Radiology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, 1111 Xianxia Road, Shanghai, 200336, People's Republic of China
| | - Yahui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Xiaoxin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Mingxuan Feng
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China
| | - Jun Ma
- Department of Interventional Radiology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, 1111 Xianxia Road, Shanghai, 200336, People's Republic of China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Xiaomei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Fang Fang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| | - Mingyi Shang
- Department of Interventional Radiology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, 1111 Xianxia Road, Shanghai, 200336, People's Republic of China.
| | - Shuheng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
32
|
Xie S, Liu G, Huang J, Hu HB, Jiang W. miR-210 promotes lung adenocarcinoma proliferation, migration, and invasion by targeting lysyl oxidase-like 4. J Cell Physiol 2019; 234:14050-14057. [PMID: 30633357 DOI: 10.1002/jcp.28093] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/07/2018] [Indexed: 12/31/2022]
Abstract
Accumulating evidence has revealed that various microRNAs are deregulated and involved in lung cancer development and metastasis. miR-210 is implicated in several cancer progression. However, the detailed biological function and role of miR-210 in lung adenocarcinoma remains unclear. Our current study was aimed to investigate the mechanism of miR-210 in lung adenocarcinoma progression. We observed that miR-210 was significantly upregulated in lung cancer cell lines (A549 and H1650) in comparison to BEAS-2B cells. In addition, we found that miR-210 was greatly elevated in lung adenocarcinoma tissues. Then, it was shown that overexpression of miR-210 was able to promote lung cancer cell proliferation and colony formation ability while inhibitors of miR-210 exhibited a reversed phenomenon. Subsequently, A549 and H1650 cell migration and invasion capacity were obviously restrained by miR-210 inhibition whereas induced by miR-210 mimics. Lysyl oxidase-like 4 (LOXL4), a member of the secreted copper-dependent amine oxidases has been found to be increased or decreased in different cancer types. Here, we confirmed that LOXL4 could serve as a downstream target of miR-210 and miR-210 promoted lung cancer progression via targeting LOXL4. In A549 and H1650 cells, knockdown of LOXL4 dramatically repressed lung cancer cell proliferation, migration, and invasion. In conclusion, our study implied that miR-210 might indicate a new perspective for lung cancer.
Collapse
Affiliation(s)
- Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaoli Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Huang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hai-Bo Hu
- Department of Thoracic Surgery, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Kielosto M, Eriksson J, Nummela P, Yin M, Hölttä E. Divergent roles of lysyl oxidase family members in ornithine decarboxylase- and RAS-transformed mouse fibroblasts and human melanoma cells. Oncotarget 2018; 9:37733-37752. [PMID: 30701028 PMCID: PMC6340875 DOI: 10.18632/oncotarget.26508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
We have previously shown that proto-oncoprotein c-Jun is activated in ornithine decarboxylase (ODC)- and RAS-transformed mouse fibroblasts, and that the transformed morphology of these cells can be reversed by expressing the transactivation domain deletion mutant of c-Jun (TAM67). Here, we found that lysyl oxidase (Lox), encoding an extracellular matrix-modifying enzyme, is downregulated in a c-Jun-dependent manner in ODC-transformed fibroblasts (Odc cells). In addition to Lox, the Lox family members Lox-like 1 and 3 (Loxl1 and Loxl3) were found to be downregulated in Odc as well as in RAS-transformed fibroblasts (E4), whereas Lox-like 4 (Loxl4) was upregulated in Odc and downregulated in E4 cells compared to normal N1 fibroblasts. Tetracycline-regulatable LOX re-expression in Odc cells led to inhibition of cell growth and invasion in three-dimensional Matrigel in an activity-independent manner. On the contrary, LOX and especially LOXL2, LOXL3, and LOXL4 were found to be upregulated in several human melanoma cell lines, and LOX inhibitor B-aminopropionitrile inhibited the invasive growth of these cells particularly when co-cultured with fibroblasts in Matrigel. Knocking down the expression of LOX and especially LOXL2 in melanoma cells almost completely abrogated the invasive growth capability. Further, LOXL2 was significantly upregulated in clinical human primary melanomas compared to benign nevi, and high expression of LOXL2 in primary melanomas was associated with formation of metastases and shorter survival of patients. Thus, our studies reveal that inactive pro-LOX (together with Lox propeptide) functions as a tumor suppressor in ODC- and RAS-transformed murine fibroblasts by inhibiting cell growth and invasion, and active LOX and LOXL2 as tumor promoters in human melanoma cells by promoting their invasive growth.
Collapse
Affiliation(s)
- Mari Kielosto
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Johanna Eriksson
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Pirjo Nummela
- Department of Pathology, University of Helsinki, Helsinki, Finland.,Current address: University of Helsinki, Genome-Scale Biology Research Program, Helsinki, Finland
| | - Miao Yin
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Erkki Hölttä
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Dynamic matrisome: ECM remodeling factors licensing cancer progression and metastasis. Biochim Biophys Acta Rev Cancer 2018; 1870:207-228. [DOI: 10.1016/j.bbcan.2018.09.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 01/04/2023]
|
35
|
CXCL5, the upregulated chemokine in patients with uterine cervix cancer, in vivo and in vitro contributes to oncogenic potential of Hela uterine cervix cancer cells. Biomed Pharmacother 2018; 107:1496-1504. [PMID: 30257367 DOI: 10.1016/j.biopha.2018.08.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 11/23/2022] Open
Abstract
CXCL5 is showed a surprisingly elevated profile and implicated in tumorigenesis in several tumors. However, the expression and function of CXCL5 in uterine cervix cancer (UCC) remain largely unknown. The current study aimed to elucidate the expression pattern of CXCL5 in human UCC tissues and Hela cervix cancer cell, as well as its functions in Hela cells. Our data showed that CXCL5 and its receptor CXCR2 were expressed by Hela uterine cervix cancer cells. CXCL5 was upregulated in UCC tissues, and its overexpression was positively correlated with age, but did not correlate with clinical stages and tumor infiltration. Exogenous administration of CXCL5 and CXCL5 overexpression contributed to proliferation and migration activities of Hela cells in vitro, consistent with this, CXCL5 overexpression also promoted growth of Hela cells in a nude mouse xenograft model. At the gene level, CXCL5 overexpression regulated the expression of tumor-related genes including ERK, p-ERK, AKT, p-AKT, DIABOL, NUMB, NDRG3 and CXCR2. Taken together, CXCL5 may contribute to a dominant role in UCC progression and sever as a potential molecular therapeutic target for UCC.
Collapse
|
36
|
Hajdú I, Kardos J, Major B, Fabó G, Lőrincz Z, Cseh S, Dormán G. Inhibition of the LOX enzyme family members with old and new ligands. Selectivity analysis revisited. Bioorg Med Chem Lett 2018; 28:3113-3118. [PMID: 30098867 DOI: 10.1016/j.bmcl.2018.07.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 01/08/2023]
Abstract
Lysyl oxidase (LOX) enzymes as potential drug targets maintain constant attention in the therapy of fibrosis, cancer and metastasis. In order to measure the inhibitory activity of small molecules on the LOX enzyme family members a fluorometric activity screening method was developed. During assay validation, previously reported non-selective small inhibitor molecules (BAPN, MCP-1, thiram, disulfiram) were investigated on all of the major LOX enzymes. We confirmed that MCP-1, thiram, disulfiram are in fact pan-inhibitors, while BAPN inhibits only LOX-like enzymes (preferably LOX-like-protein-2, LOXL2) in contrast to the previous reports. We measured the LOX inhibitory profile of a small targeted library generated by 2D ligand-based chemoinformatics methods. Ten hits (10.4% hit rate) were identified, and the compounds showed distinct activity profiles. Potential inhibitors were also identified for LOX-like-protein-3 (LOXL3) and LOX-like-protein-4 (LOXL4), that are considered as emerging drug targets in the therapy of melanoma and gastric cancer.
Collapse
Affiliation(s)
- István Hajdú
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary; Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - József Kardos
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117 Budapest, Hungary
| | - Balázs Major
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Gabriella Fabó
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary
| | - Zsolt Lőrincz
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary
| | - Sándor Cseh
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary
| | - György Dormán
- TargetEx Ltd., Madách Imre utca 31/2, H-2120 Dunakeszi, Hungary.
| |
Collapse
|
37
|
Mohammadi H, Sahai E. Mechanisms and impact of altered tumour mechanics. Nat Cell Biol 2018; 20:766-774. [PMID: 29950570 DOI: 10.1038/s41556-018-0131-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
Abstract
The physical characteristics of tumours are intricately linked to the tumour phenotype and difficulties during treatment. Many factors contribute to the increased stiffness of tumours; from increased matrix deposition, matrix remodelling by forces from cancer cells and stromal fibroblasts, matrix crosslinking, increased cellularity, and the build-up of both solid and interstitial pressure. Increased stiffness then feeds back to increase tumour invasiveness and reduce therapy efficacy. Increased understanding of this interplay is offering new therapeutic avenues.
Collapse
|
38
|
Chen J, Ren J, Loo WTY, Hao L, Wang M. Lysyl oxidases expression and histopathological changes of the diabetic rat nephron. Mol Med Rep 2017; 17:2431-2441. [PMID: 29207131 PMCID: PMC5783488 DOI: 10.3892/mmr.2017.8182] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 11/06/2017] [Indexed: 02/05/2023] Open
Abstract
Diabetic nephropathy (DN) is a major complication of diabetes, the accumulation of extracellular matrix (ECM) is considered an indication of nephropathological changes. Lysyl oxidases (LOXs) are also associated with ECM. However, the majority of studies on LOXs have focused on their potential role in renal fibrogenesis and there has no examination of LOXs expression or the correlation with histopathological changes of DN, including glomerular basement membrane (GBM) thickening and glomerulosclerosis. In this study, the association between histological changes and LOXs was explored using a type 2 diabetes model of male Zucker diabetic fatty rats. The expression of LOX and lysyl oxidase-like 1 to 3 (LOXL1 to 3) levels were evaluated by immunohistochemical staining. The expression levels of LOX and LOXL2 in the kidney tissue in the diabetic group were significantly higher compared with those of the control group, but LOXL1 and LOXL3 expression levels were not significantly different between the two groups. These results indicated that LOXL2 and LOX may be critical factors involved in the progression of DN.
Collapse
Affiliation(s)
- Jun Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jie Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wings T Y Loo
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, P.R. China
| | - Liang Hao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Min Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
39
|
Li R, Zhuang C, Jiang S, Du N, Zhao W, Tu L, Cao H, Zhang Z, Chen X. ITGBL1 Predicts a Poor Prognosis and Correlates EMT Phenotype in Gastric Cancer. J Cancer 2017; 8:3764-3773. [PMID: 29151964 PMCID: PMC5688930 DOI: 10.7150/jca.20900] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/14/2017] [Indexed: 12/25/2022] Open
Abstract
Integrin, beta-like 1 (ITGBL1), a β-integrin-related extracellular matrix protein, was found more commonly up-regulated in gastric cancer (GC) by screening and analyzing Gene Expression Omnibus (GEO) and Oncomine databases, reminding us to explore its prognostic significance in GC. In our current study, we observed that ITGBL1 expression was significantly up-regulated in GC compared with normal controls in clinical specimens. In addition, elevated ITGBL1 expression was positively correlated with patients' tumor-node-metastasis (TNM) stage and distant metastasis. Kaplan-Meier analysis indicated that high ITGBL1 expression was significantly associated with shorter survival times in GC patients. Multivariate Cox regression analysis confirmed ITGBL1 expression as an independent prognostic factor in GC. Gene set enrichment analysis (GSEA) of multiple GEO datasets revealed a close relationship between ITGBL1 expression and the KRAS/epithelial-mesenchymal transition (EMT) signaling pathway. In conclusion, these data provide evidences that ITGBL1 is a potential predictor and may be involved in cancer cell invasion and metastasis via inducing EMT, and the ITGBL1-related pathways may represent a novel therapeutic strategy for treatment of GC.
Collapse
Affiliation(s)
- Rongkun Li
- Department of Oncology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, PR China
| | - Chun Zhuang
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, PR China
| | - Shuheng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Nan Du
- Department of Oncology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, PR China
| | - Wenyi Zhao
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, PR China
| | - Lin Tu
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, PR China
| | - Hui Cao
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, PR China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Xiaofei Chen
- Department of Oncology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, PR China
| |
Collapse
|
40
|
Vitte J, Gao F, Coppola G, Judkins AR, Giovannini M. Timing of Smarcb1 and Nf2 inactivation determines schwannoma versus rhabdoid tumor development. Nat Commun 2017; 8:300. [PMID: 28824165 PMCID: PMC5563506 DOI: 10.1038/s41467-017-00346-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 06/23/2017] [Indexed: 02/06/2023] Open
Abstract
Germline mutations of the SMARCB1 gene predispose to two distinct tumor syndromes: rhabdoid tumor predisposition syndrome, with malignant pediatric tumors mostly developing in brain and kidney, and familial schwannomatosis, with adulthood benign tumors involving cranial and peripheral nerves. The mechanisms by which SMARCB1 germline mutations predispose to rhabdoid tumors versus schwannomas are still unknown. Here, to understand the origin of these two types of SMARCB1-associated tumors, we generated different tissue- and developmental stage-specific conditional knockout mice carrying Smarcb1 and/or Nf2 deletion. Smarcb1 loss in early neural crest was necessary to initiate tumorigenesis in the cranial nerves and meninges with typical histological features and molecular profiles of human rhabdoid tumors. By inducing Smarcb1 loss at later developmental stage in the Schwann cell lineage, in addition to biallelic Nf2 gene inactivation, we generated the first mouse model developing schwannomas with the same underlying gene mutations found in schwannomatosis patients. SMARCB1 mutations predispose to rhabdoid tumors and schwannomas but the mechanisms underlying the tumor type specificity are unknown. Here the authors present new mouse models and show that early Smarcb1 loss causes rhabdoid tumors whereas loss at later stages combined with Nf2 gene inactivation causes shwannomas.
Collapse
Affiliation(s)
- Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Fuying Gao
- Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Giovanni Coppola
- Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Alexander R Judkins
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
41
|
Lysyl Oxidase and the Tumor Microenvironment. Int J Mol Sci 2016; 18:ijms18010062. [PMID: 28036074 PMCID: PMC5297697 DOI: 10.3390/ijms18010062] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 12/20/2016] [Accepted: 12/23/2016] [Indexed: 12/14/2022] Open
Abstract
The lysyl oxidase (LOX) family of oxidases contains a group of extracellular copper-dependent enzymes that catalyze the cross-linking of collagen and elastin by oxidation, thus maintaining the rigidity and structural stability of the extracellular matrix (ECM). Aberrant expression or activation of LOX alters the cellular microenvironment, leading to many diseases, including atherosclerosis, tissue fibrosis, and cancer. Recently, a number of studies have shown that LOX is overexpressed in most cancers and that it is involved in the regulation of tumor progression and metastasis. In contrast, a few reports have also indicated the tumor-suppressing role of LOX. In this short review, we discuss recent research on the correlations between LOX and cancer. Further, the role of LOX in tumor microenvironment remodeling, tumorigenesis, and metastasis and the underlying mechanisms have also been elucidated.
Collapse
|
42
|
Añazco C, Delgado-López F, Araya P, González I, Morales E, Pérez-Castro R, Romero J, Rojas A. Lysyl oxidase isoforms in gastric cancer. Biomark Med 2016; 10:987-998. [PMID: 27564724 DOI: 10.2217/bmm-2016-0075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is the fifth most frequent cancer in the world and shows the highest incidence in Latin America and Asia. An increasing amount of evidence demonstrates that lysyl oxidase isoforms, a group of extracellular matrix crosslinking enzymes, should be considered as potential biomarkers and therapeutic targets in GC. In this review, we focus on the expression levels of lysyl oxidase isoforms, its functions and the clinical implications in GC. Finding novel proteins related to the processing of these extracellular matrix enzymes might be helpful in the design of new therapies, which, in combination with classic pharmacology, could be used to delay the progress of this aggressive cancer and offer a wider temporal window for clinical intervention.
Collapse
Affiliation(s)
- Carolina Añazco
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | | | - Paulina Araya
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Ileana González
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Erik Morales
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
- Pathology Department, Regional Hospital of Talca, Talca, Chile
| | - Ramón Pérez-Castro
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Jacqueline Romero
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| |
Collapse
|
43
|
Hsu YL, Hung JY, Liang YY, Lin YS, Tsai MJ, Chou SH, Lu CY, Kuo PL. S100P interacts with integrin α7 and increases cancer cell migration and invasion in lung cancer. Oncotarget 2016; 6:29585-98. [PMID: 26320193 PMCID: PMC4745748 DOI: 10.18632/oncotarget.4987] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/16/2015] [Indexed: 01/11/2023] Open
Abstract
S100P, a Ca2+ binding protein, has been shown to be overexpressed in various cancers. However, its functional character in lung cancer remains largely unknown. In this study, we show that S100P increases cancer migration, invasion and metastasis in lung cancer cells. Ectopic expression of S100P increases migration, invasion and EMT in less invasive CL1-0 lung cancer cells. Conversely, knockdown of S100P suppressed migration and invasion, and caused a reversion of EMT in highly invasive lung cancer cells. These effects were transduced by increasing the interaction of S100P with integrin α7, which activated focal adhesion kinase (FAK) and AKT. Blocking FAK significantly decreased S100P-induced migration by decreasing Src and AKT activation, whereas inhibiting AKT reduced S100P upregulation on ZEB1 expression. Further study has indicated that S100P knockdown prevents the spread of highly metastatic human lung cancer in animal models. This study therefore suggests that S100P represents a critical activator of lung cancer metastasis. Detection and targeted treatment of S100P-expressing cancer is an attractive therapeutic strategy in treating lung cancer.
Collapse
Affiliation(s)
- Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jen-Yu Hung
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yung-Yu Liang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shiuan Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Ju Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shah-Hwa Chou
- Division of Chest Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
44
|
Trackman PC. Lysyl Oxidase Isoforms and Potential Therapeutic Opportunities for Fibrosis and Cancer. Expert Opin Ther Targets 2016; 20:935-45. [PMID: 26848785 DOI: 10.1517/14728222.2016.1151003] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The lysyl oxidase family of enzymes is classically known as being required for connective tissue maturation by oxidizing lysine residues in elastin and lysine and hydroxylysine residues in collagen precursors. The resulting aldehydes then participate in cross-link formation, which is required for normal connective tissue integrity. These enzymes have biological functions that extend beyond this fundamental biosynthetic role, with contributions to angiogenesis, cell proliferation, and cell differentiation. Dysregulation of lysyl oxidases occurs in multiple pathologies including fibrosis, primary and metastatic cancers, and complications of diabetes in a variety of tissues. AREAS COVERED This review summarizes the major findings of novel roles for lysyl oxidases in pathologies, and highlights some of the potential therapeutic approaches that are in development and which stem from these new findings. EXPERT OPINION Fundamental questions remain regarding the mechanisms of novel biological functions of this family of proteins, and regarding functions that are independent of their catalytic enzyme activity. However, progress is underway in the development of isoform-specific pharmacologic inhibitors, potential therapeutic antibodies and gaining an increased understanding of both tumor suppressor and metastasis promotion activities. Ultimately, this is likely to lead to novel therapeutic agents.
Collapse
Affiliation(s)
- Philip C Trackman
- a Department of Molecular and Cell Biology , Boston University, Henry M. Goldman School of Dental Medicine , Boston , MA , USA
| |
Collapse
|
45
|
Yilmaz M, Suer I, Karatas OF, Cansiz H, Ozen M. Differential expression of LOXL4 in normal and tumour tissue samples of laryngeal squamous cell carcinoma. Clin Otolaryngol 2016; 41:206-10. [PMID: 26138381 DOI: 10.1111/coa.12498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Head and neck cancer is the sixth most common cancer in worldwide with an increasing incidence rate in recent years. LOXL4 is expressed in several tissues, and its expression has been shown to display a significant correlation with local lymph node metastasis. In this study, we aimed to explore the LOXL4 expression level in metastatic and non-metastatic LSCC tissues and to determine its prognostic significance. STUDY DESIGN Basic science research study. SETTING University hospital. PARTICIPANTS A total of 40 patients were included in the study. MAIN OUTCOME MEASURE LOXL4 expression status in metastatic, non-metastatic LSCC and normal tissue samples was investigated by quantitative reverse-transcription PCR. RESULTS We demonstrated that LOXL4 was significantly overexpressed in LSCC tumour tissue samples in comparison with the corresponding normal tissues (P < 0.001); however, no significant relationship has been found between LOXL4 expression and either the metastatic potential or the T classification of the specimens. CONCLUSIONS Although expression of LOXL4 is not statistically associated with neck metastases, we showed that LOXL4 expression significantly increased in laryngeal cancer.
Collapse
Affiliation(s)
- M Yilmaz
- Department of Otorhinolaryngology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - I Suer
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
| | - O F Karatas
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey.,Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, Turkey
| | - H Cansiz
- Department of Otorhinolaryngology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - M Ozen
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey.,Department of Molecular Biology and Genetics, Biruni University, Istanbul, Turkey.,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
46
|
Inhibition of Lysyl Oxidase and Lysyl Oxidase-Like Enzymes Has Tumour-Promoting and Tumour-Suppressing Roles in Experimental Prostate Cancer. Sci Rep 2016; 6:19608. [PMID: 26804196 PMCID: PMC4726263 DOI: 10.1038/srep19608] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 12/07/2015] [Indexed: 12/23/2022] Open
Abstract
Lysyl oxidase (LOX) and LOX-like (LOXL) enzymes are key players in extracellular matrix deposition and maturation. LOX promote tumour progression and metastasis, but it may also have tumour-inhibitory effects. Here we show that orthotopic implantation of rat prostate AT-1 tumour cells increased LOX and LOXLs mRNA expressions in the tumour and in the surrounding non-malignant prostate tissue. Inhibition of LOX enzymes, using Beta-aminopropionitrile (BAPN), initiated before implantation of AT-1 cells, reduced tumour growth. Conversely, treatment that was started after the tumours were established resulted in unaffected or increased tumour growth. Moreover, treatment with BAPN did not suppress the formation of spontaneous lymph node metastases, or lung tumour burden, when tumour cells were injected intravenously. A temporal decrease in collagen fibre content, which is a target for LOX, was observed in tumours and in the tumour-adjacent prostate tissue. This may explain why early BAPN treatment is more effective in inhibiting tumour growth compared to treatment initiated later. Our data suggest that the enzymatic function of the LOX family is context-dependent, with both tumour-suppressing and tumour-promoting properties in prostate cancer. Further investigations are needed to understand the circumstances under which LOX inhibition may be used as a therapeutic target for cancer patients.
Collapse
|
47
|
Cho A, Howell VM, Colvin EK. The Extracellular Matrix in Epithelial Ovarian Cancer - A Piece of a Puzzle. Front Oncol 2015; 5:245. [PMID: 26579497 PMCID: PMC4629462 DOI: 10.3389/fonc.2015.00245] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/15/2015] [Indexed: 02/04/2023] Open
Abstract
Epithelial ovarian cancer is the fifth leading cause of cancer-related deaths in women and the most lethal gynecological malignancy. Extracellular matrix (ECM) is an integral component of both the normal and tumor microenvironment. ECM composition varies between tissues and is crucial for maintaining normal function and homeostasis. Dysregulation and aberrant deposition or loss of ECM components is implicated in ovarian cancer progression. The mechanisms by which tumor cells induce ECM remodeling to promote a malignant phenotype are yet to be elucidated. A thorough understanding of the role of the ECM in ovarian cancer is needed for the development of effective biomarkers and new therapies.
Collapse
Affiliation(s)
- Angela Cho
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, NSW, Australia
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, Australia
| | - Viive M. Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Emily K. Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
48
|
Emerging roles of focal adhesion kinase in cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:690690. [PMID: 25918719 PMCID: PMC4396139 DOI: 10.1155/2015/690690] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/20/2015] [Indexed: 12/13/2022]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic nonreceptor tyrosine kinase that enables activation by growth factor receptors or integrins in various types of human cancers. The kinase-dependent and kinase-independent scaffolding functions of FAK modulate the authentic signaling and fundamental functions not only in cancer cells but also in tumor microenvironment to facilitate cancer progression and metastasis. The overexpression and activation of FAK are usually investigated in primary or metastatic cancers and correlated with the poor clinical outcome, highlighting FAK as a potential prognostic marker and anticancer target. Small molecule inhibitors targeting FAK kinase activity or FAK-scaffolding functions impair cancer development in preclinical or clinical trials. In this review, we give an overview for FAK signaling in cancer cells as well as tumor microenvironment that provides new strategies for the invention of cancer development and malignancy.
Collapse
|