1
|
Hidalgo I, Sorolla MA, Sorolla A, Salud A, Parisi E. Secreted Phospholipases A2: Drivers of Inflammation and Cancer. Int J Mol Sci 2024; 25:12408. [PMID: 39596471 PMCID: PMC11594849 DOI: 10.3390/ijms252212408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Secreted phospholipase 2 (sPLA2) is the largest family of phospholipase A2 (PLA2) enzymes with 11 mammalian isoforms. Each sPLA2 exhibits different localizations and specific properties, being involved in a very wide spectrum of biological processes. The enzymatic activity of sPLA2 has been well described; however, recent findings have shown that they could regulate different signaling pathways by acting directly as ligands. Arachidonic acid (AA) and its derivatives are produced by sPLA2 in collaboration with other molecules in the extracellular space, making important impacts on the cellular environment, being especially relevant in the contexts of immunity and cancer. For these reasons, this review focuses on sPLA2 functions in processes such as the promotion of EMT, angiogenesis, and immunomodulation in the context of tumor initiation and progression. Finally, we will also describe how this knowledge has been applied in the search for new sPLA2 inhibitory compounds that can be used for cancer treatment.
Collapse
Affiliation(s)
- Ivan Hidalgo
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Maria Alba Sorolla
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Anabel Sorolla
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Antonieta Salud
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
- Department of Medical Oncology, Arnau de Vilanova University Hospital (HUAV), 25198 Lleida, Spain
- Department of Medicine, University of Lleida, 25198 Lleida, Spain
| | - Eva Parisi
- Research Group of Cancer Biomarkers, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
- Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain
| |
Collapse
|
2
|
Tufail M, Wu C, Hussain MS. Dietary, addictive and habitual factors, and risk of colorectal cancer. Nutrition 2024; 120:112334. [PMID: 38271761 DOI: 10.1016/j.nut.2023.112334] [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: 07/20/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND In Pakistan, the incidence of colorectal cancer (CRC) has sharply increased in recent years. Although several studies have reported global risk factors for CRC, no study has been conducted in Khyber Pakhtunkhwa (KPK), Pakistan, to investigate the risk factors associated with the increased CRC burden in this population. OBJECTIVES Therefore, we conducted a clinical survey using a case-control study design to explore the risk factors associatd with CRC. METHODS In the present study, one control was enrolled for each case. Both cases and controls were asked to complete a questionnaire to gather data. We analyzed all data using SPSS. RESULTS Our study found that certain dietary factors, such as consuming fast food (OR: 3.0; P = 0.0001) and reusing ghee (OR: 2.45; P = 0.0001) and oil (OR: 4.30; P = 0.0001), increase the risk of CRC. Additionally, use of tobacco products like smoking cigarettes (OR: 1.91; P = 0.0001) and using snuff (OR: 3.72; P = 0.0001) significantly increases the risk of CRC. Certain habitual factors, including binge eating (OR: 2.42; P = 0.0001) and spending excessive time watching TV (OR: 1.98; P = 0.0001), also increase the odds of developing CRC. However, our study also identified some protective factors against CRC, such as consuming vegetables (OR: .41; P = 0.0001), developing healthy eating habits (OR: .61; P = 0.0001), and maintaining regular sleeping patterns (OR: .45; P = 0.0001). CONCLUSION Given these findings, targeted health education is necessary to prevent the increase in CRC in this area. We also recommend developing and enforcing appropriate control guidelines for cancer risk factors to curb the incidence of CRC.
Collapse
Affiliation(s)
- Muhammad Tufail
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China.
| | - Changxin Wu
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| |
Collapse
|
3
|
Zhang J, Shi L, Duan J, Li M, Li C. Proteomic detection of COX-2 pathway-related factors in patients with adenomyosis. PeerJ 2024; 12:e16784. [PMID: 38239300 PMCID: PMC10795527 DOI: 10.7717/peerj.16784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/19/2023] [Indexed: 01/22/2024] Open
Abstract
Background Investigating the relationship between cyclooxygenase-2 (COX-2) pathway-related factors and clinical features in patients with adenomyosis by proteomics could provide potential therapeutic targets. Methods This study recruited 40 patients undergoing surgical hysterectomy and pathological diagnosis of adenomyosis, collected ectopic endometrial specimens, and recorded clinical data. The expression levels of COX-2 in ectopic uterus lesions were detected using the immunohistochemical (IHC) SP method. The 40 samples were then divided into a COX-2 low or high expression group. Five samples with the most typical expression levels were selected from each of the two groups and the differential proteins between the two groups were identified using label-free quantitative proteomics. WW domain-binding protein 2 (WBP2), interferon induced transmembrane protein 3 (IFITM3), and secreted frizzled-related protein 4 (SFRP4) were selected for further verification, and their relationships with COX-2 and clinical characteristics were analyzed. Results There were statistically significant differences in the expression of WBP2, IFITM3, and SFRP4 between the COX-2 low and high expression groups (P < 0.01). The expressions of COX-2, IFITM3, and SFRP4 were significantly correlated with dysmenorrhea between the two groups (P < 0.05), but not with uterine size or menstrual volume (P > 0.05). However, there was no significant correlation between the expression of WBP2 and dysmenorrhea, uterine size, and menstruation volume in both the high expression and low expression groups (P > 0.05). Conclusions COX-2, IFITM3, SFRP4, and WBP2 may be involved in the pathogenesis of adenomyosis. COX-2, IFITM3, and SFRP4 may serve as potential molecular biomarkers or therapeutic targets in dysmenorrhea in patients with early adenomyosis.
Collapse
Affiliation(s)
- Jihua Zhang
- Department of Gynecology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Luying Shi
- Department of Gynecology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingya Duan
- Department of Gynecology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Minmin Li
- Department of Gynecology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Canyu Li
- Department of Gynecology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Cai X, Lv Y, Pan J, Cao Z, Zhang J, Li Y, Zheng H. CBX8 Promotes Epithelial-mesenchymal Transition, Migration, and Invasion of Lung Cancer through Wnt/β-catenin Signaling Pathway. Curr Protein Pept Sci 2024; 25:386-393. [PMID: 38265409 DOI: 10.2174/0113892037273375231204080906] [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: 07/21/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Lung cancer (LC) is primarily responsible for cancer-related deaths worldwide. Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells acquire mesenchymal features and is associated with the development of tumors. CBX8, a member of the PcG protein family, plays a critical role in various cancers, containing LC. However, specific regulatory mechanisms of CBX8 in LC progression are not fully understood. This study aimed to investigate the regulatory role of CBX8 in LC progression. METHODS Bioinformatics was used to analyze the relationship between CBX8 level and tumor and the enrichment pathway of CBX8 enrichment. qRT-PCR was used to detect the differential expression of CBX8 in LC cells and normal lung epithelial cells. The effects of knockdown or overexpression of CBX8 on the proliferation, migration and invasion of LC cells were evaluated by CCK- -8 assay and Transwell assay, and the levels of proteins associated with the EMT pathway and Wnt/ β-catenin signaling pathway were detected by western blot. RESULTS Bioinformatics analysis revealed that CBX8 was highly expressed in LC and enriched on the Wnt/β-catenin signaling pathway. The expression level of CBX8 was significantly elevated in LC cells. Knockdown of CBX8 significantly inhibited cell proliferation, migration and invasion, and decreased the expression levels of EMT-related proteins and Wnt/β-catenin pathway-related proteins. Conversely, overexpression of CBX8 promoted cell proliferation, migration and invasion, and increased the expression levels of EMT-related proteins and Wnt/β-catenin pathway-related proteins. The Wnt inhibitor IWP-4 alleviated the effects produced by overexpression of CBX8. CONCLUSION Collectively, these data demonstrated that CBX8 induced EMT through Wnt/β-- catenin signaling, driving migration and invasion of LC cells.
Collapse
Affiliation(s)
- Xiaoping Cai
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Yuankai Lv
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Jiongwei Pan
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Zhuo Cao
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Junzhi Zhang
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Yuling Li
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Hao Zheng
- Department of Respiratory, Six affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| |
Collapse
|
5
|
Liu Y, Lu L, Yang H, Wu X, Luo X, Shen J, Xiao Z, Zhao Y, Du F, Chen Y, Deng S, Cho CH, Li Q, Li X, Li W, Wang F, Sun Y, Gu L, Chen M, Li M. Dysregulation of immunity by cigarette smoking promotes inflammation and cancer: A review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 339:122730. [PMID: 37838314 DOI: 10.1016/j.envpol.2023.122730] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
Smoking is a serious global health issue. Cigarette smoking contains over 7000 different chemicals. The main harmful components include nicotine, acrolein, aromatic hydrocarbons and heavy metals, which play the key role for cigarette-induced inflammation and carcinogenesis. Growing evidences show that cigarette smoking and its components exert a remarkable impact on regulation of immunity and dysregulated immunity promotes inflammation and cancer. Therefore, this comprehensive and up-to-date review covers four interrelated topics, including cigarette smoking, inflammation, cancer and immune system. The known harmful chemicals from cigarette smoking were summarized. Importantly, we discussed in depth the impact of cigarette smoking on the formation of inflammatory or tumor microenvironment, primarily by affecting immune effector cells, such as macrophages, neutrophils, and T lymphocytes. Furthermore, the main molecular mechanisms by which cigarette smoking induces inflammation and cancer, including changes in epigenetics, DNA damage and others were further summarized. This article will contribute to a better understanding of the impact of cigarette smoking on inducing inflammation and cancer.
Collapse
Affiliation(s)
- Yubin Liu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Lan Lu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Huan Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xinyue Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Qianxiu Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China.
| |
Collapse
|
6
|
Hu QR, Huang QX, Hong H, Pan Y, Luo T, Li J, Deng ZY, Chen F. Ginsenoside Rh2 and its octyl ester derivative inhibited invasion and metastasis of hepatocellular carcinoma via the c-Jun/COX2/PGE2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155131. [PMID: 37806155 DOI: 10.1016/j.phymed.2023.155131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Liver cancer is a topical global health issue. The treatment of liver cancer meets significant challenges in the high recurrence rate and invasive incidence. Therefore, the treatment strategies that target epithelial-mesenchymal transition (EMT) induced by cyclooxygenase 2 (COX2)/ prostaglandin E2 (PGE2) pathway have become epidemic. Ginsenoside Rh2 has been proved to inhibit the EMT. However, the underlying mechanisms remain unclear. Moreover, the octyl ester derivative of Rh2 (Rh2-O) exhibited superior anti-proliferative and immunomodulatory effects than Rh2 in our previous researches, which indicated that Rh2-O might also exert inhibitory effects on invasion and metastasis. PURPOSE The aim of current study is to explore the inhibitory effects of Rh2 and Rh2-O on invasion and metastasis of hepatocellular carcinoma, and to investigate whether these effects are dependent on the c-Jun/COX2/PGE2 pathway. STUDY DESIGN The Huh-7 liver cancer cells and the H22 tumor-bearing mice were treated with Rh2 and Rh2-O. METHOD In this paper, the inhibitory effects of Rh2 and Rh2-O on invasion and metastasis were tested by wound healing, trans-well assay and tumor-bearing mice, and the involvement of c-Jun/COX2/PGE2 pathway were verified by exogenous PGE2, activation of COX2 and overexpression of c-Jun. RESULTS The results showed that Rh2 and Rh2-O could efficiently inhibit the invasion and metastasis in a dose-dependent manner (p < 0.05). And the Rh2-O showed stronger effects than Rh2. Moreover, the exogenous PGE2, activation of COX2 by exogenous LPS and the overexpression of c-Jun by transfection all reversed the inhibitory effects of Rh2 and Rh2-O on metastasis or EMT (p < 0.05). CONCLUSION Rh2 and Rh2-O could inhibit the invasion and metastasis of hepatocellular carcinoma via restraining the EMT, which was mediated by c-Jun/COX2/PGE2 pathway.
Collapse
Affiliation(s)
- Qi-Rui Hu
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Qing-Xin Huang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi 330000, China
| | - Huan Hong
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi 330000, China
| | - Yao Pan
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi 330000, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Ze-Yuan Deng
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Fang Chen
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi 330000, China.
| |
Collapse
|
7
|
Papapostolou I, Ross-Kaschitza D, Bochen F, Peinelt C, Maldifassi MC. Contribution of the α5 nAChR Subunit and α5SNP to Nicotine-Induced Proliferation and Migration of Human Cancer Cells. Cells 2023; 12:2000. [PMID: 37566079 PMCID: PMC10417634 DOI: 10.3390/cells12152000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
Nicotine in tobacco is known to induce tumor-promoting effects and cause chemotherapy resistance through the activation of nicotinic acetylcholine receptors (nAChRs). Many studies have associated the α5 nicotinic receptor subunit (α5), and a specific polymorphism in this subunit, with (i) nicotine administration, (ii) nicotine dependence, and (iii) lung cancer. The α5 gene CHRNA5 mRNA is upregulated in several types of cancer, including lung, prostate, colorectal, and stomach cancer, and cancer severity is correlated with smoking. In this study, we investigate the contribution of α5 in the nicotine-induced cancer hallmark functions proliferation and migration, in breast, colon, and prostate cancer cells. Nine human cell lines from different origins were used to determine nAChR subunit expression levels. Then, selected breast (MCF7), colon (SW480), and prostate (DU145) cancer cell lines were used to investigate the nicotine-induced effects mediated by α5. Using pharmacological and siRNA-based experiments, we show that α5 is essential for nicotine-induced proliferation and migration. Additionally, upon downregulation of α5, nicotine-promoted expression of EMT markers and immune regulatory proteins was impaired. Moreover, the α5 polymorphism D398N (α5SNP) caused a basal increase in proliferation and migration in the DU145 cell line, and the effect was mediated through G-protein signaling. Taken together, our results indicate that nicotine-induced cancer cell proliferation and migration are mediated via α5, adding to the characterization of α5 as a putative therapeutical target.
Collapse
Affiliation(s)
| | | | | | | | - Maria Constanza Maldifassi
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland; (I.P.); (D.R.-K.); (F.B.); (C.P.)
| |
Collapse
|
8
|
Gesualdi L, Berardini M, Scicchitano BM, Castaldo C, Bizzarri M, Filippini A, Riccioli A, Schiraldi C, Ferranti F, Liguoro D, Mancini R, Ricci G, Catizone A. ERK Signaling Pathway Is Constitutively Active in NT2D1 Non-Seminoma Cells and Its Inhibition Impairs Basal and HGF-Activated Cell Proliferation. Biomedicines 2023; 11:1894. [PMID: 37509533 PMCID: PMC10377482 DOI: 10.3390/biomedicines11071894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
c-MET/hepatocyte growth factor (HGF) system deregulation is a well-known feature of malignancy in several solid tumors, and for this reason this system and its pathway have been considered as potential targets for therapeutic purposes. In previous manuscripts we reported c-MET/HGF expression and the role in testicular germ cell tumors (TGCTs) derived cell lines. We demonstrated the key role of c-Src and phosphatidylinositol 3-kinase (PI3K)/AKT adaptors in the HGF-dependent malignant behavior of the embryonal carcinoma cell line NT2D1, finding that the inhibition of these onco-adaptor proteins abrogates HGF triggered responses such as proliferation, migration, and invasion. Expanding on these previous studies, herein we investigated the role of mitogen-activated protein kinase (MAPK)/extracellular signal regulated kinase (ERK) pathways in the HGF-dependent and HGF-independent NT2D1 cells biological responses. To inhibit MAPK/ERK pathways we chose a pharmacological approach, by using U0126 inhibitor, and we analyzed cell proliferation, collective migration, and chemotaxis. The administration of U0126 together with HGF reverts the HGF-dependent activation of cell proliferation but, surprisingly, does not exert the same effect on NT2D1 cell migration. In addition, we found that the use of U0126 alone significantly promotes the acquisition of NT2D1 «migrating phenotype», while collective migration of NT2D1 cells was stimulated. Notably, the inhibition of ERK activation in the absence of HGF stimulation resulted in the activation of the AKT-mediated pathway, and this let us speculate that the paradoxical effects obtained by using U0126, which are the increase of collective migration and the acquisition of partial epithelium-mesenchyme transition (pEMT), are the result of compensatory pathways activation. These data highlight how the specific response to pathway inhibitors, should be investigated in depth before setting up therapy.
Collapse
Affiliation(s)
- Luisa Gesualdi
- Section of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Marika Berardini
- Section of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Bianca Maria Scicchitano
- Section of Histology, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
- Systems Biology Group Lab, 00161 Rome, Italy
| | - Antonio Filippini
- Section of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Anna Riccioli
- Section of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Chiara Schiraldi
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Francesca Ferranti
- Human Spaceflight and Scientific Research Unit, Italian Space Agency, 00133 Roma, Italy
| | - Domenico Liguoro
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Angela Catizone
- Section of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
9
|
R I A, Vatsyayan A, Damodaran D, Sivadas A, Van der Speeten K. Multi-omics Analysis Classifies Colorectal Cancer into Distinct Methylated Immunogenic and Angiogenic Subtypes Based on Anatomical Laterality. Indian J Surg Oncol 2023; 14:209-219. [PMID: 37359923 PMCID: PMC10284779 DOI: 10.1007/s13193-023-01760-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/25/2023] [Indexed: 06/28/2023] Open
Abstract
We employed supervised machine learning algorithms to a cohort of colorectal cancer patients from the NCI to differentiate and classify the heterogenous disease based on anatomical laterality and multi-omics stratification, in a first of its kind. Multi-omics integrative analysis shows distinct clustering of left and right colorectal cancer with disentangled representation of methylome and delineation of transcriptome and genome. We present novel multi-omics findings consistent with augmented hypermethylation of genes in right CRC, epigenomic biomarkers on the right in conjunction with immune-mediated pathway signatures, and lymphocytic invasion which unlocks unique therapeutic avenues. Contrarily, left CRC multi-omics signature is found to be marked by angiogenesis, cadherins, and epithelial-mesenchymal transition (EMT). An integrated multi-omics molecular signature of RNF217-AS1, hsa-miR-10b, and panel of FBX02, FBX06, FBX044, MAD2L2, and MIIP copy number altered genes have been found by the study. Overall survival analysis reveals genomic biomarkers ABCA13 and TTN in 852 LCRC cases, and SOX11 in 170 RCRC cases that predicts a significant survival benefit. Our study exemplifies the translational competence and robustness of machine learning in effective translational bridging of research and clinic. Supplementary Information The online version contains supplementary material available at 10.1007/s13193-023-01760-6.
Collapse
Affiliation(s)
- Anu R I
- Department of Cancer Biology and Therapeutics, MVR Cancer Center and Research Institute, Calicut, Kerala India
- Department of Clinical Biochemistry, MVR Cancer Center and Research Institute, Calicut, Kerala India
| | - Aastha Vatsyayan
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Dileep Damodaran
- Department of Surgical Oncology, MVR Cancer Center and Research Institute, Calicut, Kerala India
| | - Ambily Sivadas
- Division of Nutrition, St. John’s Research Institute, Bangalore, India
| | - Kurt Van der Speeten
- Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium
- Faculty of Medicine and Life Sciences, BIOMED Research Institute, University Hasselt, Hasselt, Belgium
| |
Collapse
|
10
|
Chen H, Tao X, Cao H, Li B, Sun Q, Wang W, Zou Y, Mu M, Tao H, Zhao Y, Ge D. Nicotine exposure exacerbates silica-induced pulmonary fibrosis via STAT3-BDNF-TrkB-mediated epithelial-mesenchymal transition in alveolar type II cells. Food Chem Toxicol 2023; 175:113694. [PMID: 36868510 DOI: 10.1016/j.fct.2023.113694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023]
Abstract
The addictive substance nicotine, found in cigarettes and some e-cigarettes, plays a vital role in pro-inflammatory and fibrotic processes. However, the part played by nicotine in the progression of silica-induced pulmonary fibrosis is poorly understood. We used mice exposed to both silica and nicotine to investigate whether nicotine synergizes with silica particles to worsen lung fibrosis. The results revealed that nicotine accelerated the development of pulmonary fibrosis in silica-injured mice by activating STAT3-BDNF-TrkB signalling. Mice with a history of exposure to nicotine showed an increase in Fgf7 expression and alveolar type II cell proliferation if they were also exposed to silica. However, newborn AT2 cells could not regenerate the alveolar structure and release pro-fibrotic factor IL-33. Moreover, activated TrkB induced the expression of p-AKT, which promotes the expression of epithelial-mesenchymal transcription factor Twist, but no Snail. In vitro assessment confirmed activation of the STAT3-BDNF-TrkB pathway in AT2 cells, exposed to nicotine plus silica. In addition, TrkB inhibitor K252a downregulated p-TrkB and the downstream p-AKT and restricted the epithelial-mesenchymal transition caused by nicotine plus silica. In conclusion, nicotine activates the STAT3-BDNF-TrkB pathway, which promotes epithelial-mesenchymal transition and exacerbates pulmonary fibrosis in mice with combined exposure to silica particles and nicotine.
Collapse
Affiliation(s)
- Haoming Chen
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Xinrong Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China.
| | - Hangbing Cao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Bing Li
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Qixian Sun
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Wenyang Wang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Yuanjie Zou
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Huihui Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Yehong Zhao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| | - Deyong Ge
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Anhui University of Science and Technology, Huainan, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|
11
|
Chen SM, Zhao CK, Yao LC, Wang LX, Ma YN, Meng L, Cai SQ, Liu CY, Qu LK, Jia YX, Shou CC. Aiphanol, a multi-targeting stilbenolignan, potently suppresses mouse lymphangiogenesis and lymphatic metastasis. Acta Pharmacol Sin 2023; 44:189-200. [PMID: 35778489 PMCID: PMC9813257 DOI: 10.1038/s41401-022-00940-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/07/2022] [Indexed: 01/18/2023]
Abstract
The high incidence of lymphatic metastasis is closely related to poor prognosis and mortality in cancers. Potent inhibitors to prevent pathological lymphangiogenesis and lymphatic spread are urgently needed. The VEGF-C-VEGFR3 pathway plays a vital role in driving lymphangiogenesis and lymph node metastasis. In addition, COX2 in tumor cells and tumor-associated macrophages (TAMs) facilitates lymphangiogenesis. We recently reported that aiphanol, a natural stilbenolignan, attenuates tumor angiogenesis by repressing VEGFR2 and COX2. In this study, we evaluated the antilymphangiogenic and antimetastatic potency of aiphanol using in vitro, ex vivo and in vivo systems. We first demonstrated that aiphanol directly bound to VEGFR3 and blocked its kinase activity with an half-maximal inhibitory concentration (IC50) value of 0.29 μM in an in vitro ADP-GloTM kinase assay. Furthermore, we showed that aiphanol (7.5-30 μM) dose-dependently counteracted VEGF-C-induced proliferation, migration and tubular formation of lymphatic endothelial cells (LECs), which was further verified in vivo. VEGFR3 knockdown markedly mitigated the inhibitory potency of aiphanol on lymphangiogenesis. In 4T1-luc breast tumor-bearing mice, oral administration of aiphanol (5 and 30 mg· kg-1 ·d-1) dose-dependently decreased lymphatic metastasis and prolonged survival time, which was associated with impaired lymphangiogenesis, angiogenesis and, interestingly, macrophage infiltration. In addition, we found that aiphanol decreased the COX2-dependent secretion of PGE2 and VEGF-C from tumor cells and macrophages. These results demonstrate that aiphanol is an appealing agent for preventing lymphangiogenesis and lymphatic dissemination by synergistically targeting VEGFR3 and inhibiting the COX2-PGE2-VEGF-C signaling axis.
Collapse
Affiliation(s)
- Shan-Mei Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chuan-Ke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Li-Cheng Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Li-Xin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yu-Nan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Laboratory Animal, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lin Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shao-Qing Cai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Cai-Yun Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Li-Ke Qu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Yan-Xing Jia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Cheng-Chao Shou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
12
|
Study on the Action Mechanism of the Yifei Jianpi Tongfu Formula in Treatment of Colorectal Cancer Lung Metastasis Based on Network Analysis, Molecular Docking, and Experimental Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6229444. [PMID: 35942366 PMCID: PMC9356795 DOI: 10.1155/2022/6229444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
Objective The lung is the second most common site of colorectal cancer (CRC) metastasis. This study aims to investigate the therapeutic effects and potential action mechanisms of Yifei Jianpi Tongfu formula (YJTF) in CRC lung metastasis in a comprehensive and systematic way by network analysis, molecular docking, and experimental verification. Methods The main ingredients in YJTF were screened from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP) and Traditional Chinese Medicine Integrated Database (TCMID), and the disease-related targets from the Online Mendelian Inheritance in Man (OMIM) and GeneCards and the compound-related targets from SwissTargetPrediction were collected. Then, Metascape was used for pathway annotation and enrichment analysis, and meanwhile, a protein-protein interaction (PPI) network was constructed. Molecular docking was carried out to investigate interactions between the active compounds and the potential targets. The in vivo effect of YJTF on CRC lung metastasis was observed in a tail vein injection mouse model. Results A total of 243 active compounds and 81 disease-related targets of YJTF were selected for analysis. The results of multiple network analysis showed that the core targets of YJTF were enriched onto various cancer-related pathways, especially focal adhesion and adherens junction. The results of molecular docking demonstrated that all core compounds (quercetin, kaempferol, luteolin, apigenin, and isorhamnetin) were capable of binding with AKT1, EGFR, SRC, ESR1, and PTGS2. Experimental validation in vivo demonstrated that YJTF combined with oxaliplatin could significantly reduce the number of lung metastases and improve the quality of life in mice. Further research suggested that YJTF inhibited CRC lung metastasis probably by modulating epithelial-to-mesenchymal transition (EMT). Conclusions According to the analysis, YJTF can be considered as an effective adjuvant therapy for CRC lung metastasis.
Collapse
|
13
|
Khare A, Burke CA, Heald B, O'Malley M, LaGuardia L, Milicia S, Cruise M, Eng C, Mankaney G. Endoscopic Findings in Patients With PTEN Hamartoma Tumor Syndrome Undergoing Surveillance. J Clin Gastroenterol 2022; 56:e183-e188. [PMID: 34231499 DOI: 10.1097/mcg.0000000000001580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/28/2021] [Indexed: 12/30/2022]
Abstract
GOALS AND BACKGROUND Phosphatase and tensin homolog hamartoma tumor syndrome (PHTS) is an inherited disorder that increases the risk for cancer in multiple organ systems, including breast, endometrial, thyroid, and the gastrointestinal tract. Surveillance is recommended however there lacks data to describe the change in polyposis phenotype and cancer incidence over surveillance. Our aim is to describe the polyposis phenotype and cancer incidence in PHTS patients undergoing endoscopic surveillance. STUDY PHTS patients, ages 17 through 89, who underwent at least 2 esophagogastroduodenoscopy (EGDs) or colonoscopies were identified. Number and sizes of polyps were noted, from which 5 categories were recreated. Incidence of colorectal and gastric cancer was evaluated. RESULTS Seventy patients were included. Patients were clustered and classified into 1 of 5 categories: no polyps, few small polyps (<1 cm, < 10 polyps), few large polyps (≥1 cm, < 10 polyps), many small polyps (<1 cm, ≥10 polyps), many large polyps (≥1 cm, ≥10 polyps). There was no significant difference in polyp number or size on EGD (P=0.47 and 0.83, respectively) or colonoscopy (P=0.49 and 0.10, respectively) over the surveillance period (4.8±3.9 y for stomach and 5.6±4.4 y for colon). The average interval between endoscopies was 28±24 months for EGDs and 29±23 months for colonoscopies. A stage II transverse colon adenocarcinoma and stage IV gastric adenocarcinoma were identified. Standardized incidence rates for gastric and colon cancers were 5427 (P=0.0002) and 353 (P=0.002), respectively. CONCLUSIONS PTHS individuals can be classified into polyposis phenotypes which do not change over an endoscopic surveillance period. Two cancers were associated with a large size polyp phenotype. Surveillance intervals should be determined by polyp size ≥1 cm and pathology.
Collapse
Affiliation(s)
| | - Carol A Burke
- Gastroenterology, Hepatology, and Nutrition
- Colorectal Surgery
- Pathology and Laboratory Medicine Institute
| | - Brandie Heald
- Center for Personalized Genetic Healthcare
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| | - Margaret O'Malley
- Colorectal Surgery
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| | - Lisa LaGuardia
- Colorectal Surgery
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| | - Susan Milicia
- Colorectal Surgery
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| | - Michael Cruise
- Pathology and Laboratory Medicine Institute
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| | - Charis Eng
- Genomic Medicine Institute
- Center for Personalized Genetic Healthcare
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| | - Gautam Mankaney
- Gastroenterology, Hepatology, and Nutrition
- Sanford R. Weiss, M.D. Center for Hereditary Colorectal Neoplasia, Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
14
|
Peerapen P, Sueksakit K, Boonmark W, Yoodee S, Thongboonkerd V. ARID1A knockdown enhances carcinogenesis features and aggressiveness of Caco-2 colon cancer cells: An in vitro cellular mechanism study. J Cancer 2022; 13:373-384. [PMID: 35069887 PMCID: PMC8771531 DOI: 10.7150/jca.65511] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/30/2021] [Indexed: 01/05/2023] Open
Abstract
Loss of ARID1A, a tumor suppressor gene, is associated with the higher grade of colorectal cancer (CRC). However, molecular and cellular mechanisms underlying the progression and aggressiveness of CRC induced by the loss of ARID1A remain poorly understood. Herein, we evaluated cellular mechanisms underlying the effects of ARID1A knockdown on the carcinogenesis features and aggressiveness of CRC cells. A human CRC cell line (Caco-2) was transfected with small interfering RNA (siRNA) specific to ARID1A (siARID1A) or scrambled (non-specific) siRNA (siControl). Cell death, proliferation, senescence, chemoresistance and invasion were then evaluated. In addition, formation of polyploid giant cancer cells (PGCCs), self-aggregation (multicellular spheroid) and secretion of an angiogenic factor, vascular endothelial growth factor (VEGF), were examined. The results showed that ARID1A knockdown led to significant decreases in cell death and senescence. On the other hand, ARID1A knockdown enhanced cell proliferation, chemoresistance and invasion. The siARID1A-transfected cells also had greater number of PGCCs and larger spheroid size and secreted greater level of VEGF compared with the siControl-transfected cells. These data, at least in part, explain the cellular mechanisms of ARID1A deficiency in carcinogenesis and aggressiveness features of CRC.
Collapse
Affiliation(s)
- Paleerath Peerapen
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kanyarat Sueksakit
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Wanida Boonmark
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sunisa Yoodee
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
15
|
The gastrointestinal microbiota in colorectal cancer cell migration and invasion. Clin Exp Metastasis 2021; 38:495-510. [PMID: 34748126 DOI: 10.1007/s10585-021-10130-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Colorectal carcinoma is the third most common cancer in developed countries and the second leading cause of cancer-related mortality. Interest in the influence of the intestinal microbiota on CRC emerged rapidly in the past few years, and the close presence of microbiota to the tumour mass creates a unique microenvironment in CRC. The gastrointestinal microbiota secrete factors that can contribute to CRC metastasis by influencing, for example, epithelial-to-mesenchymal transition. Although the role of EMT in metastasis is well-studied, mechanisms by which gastrointestinal microbiota contribute to the progression of CRC remain poorly understood. In this review, we will explore bacterial factors that contribute to the migration and invasion of colorectal carcinoma and the mechanisms involved. Bacteria involved in the induction of metastasis in primary CRC include Fusobacterium nucleatum, Enterococcus faecalis, enterotoxigenic Bacteroides fragilis, Escherichia coli and Salmonella enterica. Examples of prominent bacterial factors secreted by these bacteria include Fusobacterium adhesin A and Bacteroides fragilis Toxin. Most of these factors induce EMT-like properties in carcinoma cells and, as such, contribute to disease progression by affecting cell-cell adhesion, breakdown of the extracellular matrix and reorganisation of the cytoskeleton. It is of utmost importance to elucidate how bacterial factors promote CRC recurrence and metastasis to increase patient survival. So far, mainly animal models have been used to demonstrate this interplay between the host and microbiota. More human-based models are needed to study the mechanisms that promote migration and invasion and mimic the progression and recurrence of CRC.
Collapse
|
16
|
Gómez-Valenzuela F, Escobar E, Pérez-Tomás R, Montecinos VP. The Inflammatory Profile of the Tumor Microenvironment, Orchestrated by Cyclooxygenase-2, Promotes Epithelial-Mesenchymal Transition. Front Oncol 2021; 11:686792. [PMID: 34178680 PMCID: PMC8222670 DOI: 10.3389/fonc.2021.686792] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) corresponds to a complex and dynamic interconnection between the extracellular matrix and malignant cells and their surrounding stroma composed of immune and mesenchymal cells. The TME has constant cellular communication through cytokines that sustain an inflammatory profile, which favors tumor progression, angiogenesis, cell invasion, and metastasis. Although the epithelial-mesenchymal transition (EMT) represents a relevant metastasis-initiating event that promotes an invasive phenotype in malignant epithelial cells, its relationship with the inflammatory profile of the TME is poorly understood. Previous evidence strongly suggests that cyclooxygenase-2 (COX-2) overexpression, a pro-inflammatory enzyme related to chronic unresolved inflammation, is associated with common EMT-signaling pathways. This review article summarizes how COX-2 overexpression, within the context of the TME, orchestrates the EMT process and promotes initial metastatic-related events.
Collapse
Affiliation(s)
- Fernán Gómez-Valenzuela
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrico Escobar
- Department of Oral Pathology and Medicine, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapy - Bellvitge, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Viviana P Montecinos
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
17
|
Mao L, Yang J, Yue J, Chen Y, Zhou H, Fan D, Zhang Q, Buraschi S, Iozzo RV, Bi X. Decorin deficiency promotes epithelial-mesenchymal transition and colon cancer metastasis. Matrix Biol 2020; 95:1-14. [PMID: 33065248 DOI: 10.1016/j.matbio.2020.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 12/22/2022]
Abstract
The tumor microenvironment encompasses a complex cellular network that includes cancer-associated fibroblasts, inflammatory cells, neo-vessels, and an extracellular matrix enriched in angiogenic growth factors. Decorin is one of the main components of the tumor stroma, but it is not expressed by cancer cells. Lack of this proteoglycan correlates with down-regulation of E-cadherin and induction of β-catenin signaling. In this study, we investigated the role of a decorin-deficient tumor microenvironment in colon carcinoma progression and metastasis. We utilized an established model of colitis-associated cancer by administering Azoxymethane/Dextran sodium sulfate to adult wild-type and Dcn-/- mice. We discovered that after 12 weeks, all the animals developed intestinal tumors independently of their genotype. However, the number of intestinal neoplasms was significantly higher in the Dcn-/- microenvironment vis-à-vis wild-type mice. Mechanistically, we found that under unchallenged basal conditions, the intestinal epithelium of the Dcn-/- mice showed a significant increase in the protein levels of epithelial-mesenchymal transition associated factors including Snail, Slug, Twist, and MMP2. In comparison, in the colitis-associated cancer evoked in the Dcn-/- mice, we found that intercellular adhesion molecule 1 (ICAM-1) was also significantly increased, in parallel with epithelial-mesenchymal transition signaling pathway-related factors. Furthermore, a combined Celecoxib/decorin treatment revealed a promising therapeutic efficacy in treating human colorectal cancer cells, in decorin-deficient animals. Collectively, our results shed light on colorectal cancer progression and provide a protein-based therapy, i.e., treatment using recombinant decorin, to target the tumor microenvironment.
Collapse
Affiliation(s)
- Liping Mao
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Jinxue Yang
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Jiaxin Yue
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Yang Chen
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Hongrui Zhou
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Dongdong Fan
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Qiuhua Zhang
- Department of Pharmacology, Liaoning University of Traditional Chinese Medicine, Shenyang 110036, China
| | - Simone Buraschi
- Department of Pathology, Anatomy and Cell Biology, and Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| | - Xiuli Bi
- College of Life Science, Liaoning University, Shenyang 110036, China.
| |
Collapse
|
18
|
Water-Pipe Smoking Exposure Deregulates a Set of Genes Associated with Human Head and Neck Cancer Development and Prognosis. TOXICS 2020; 8:toxics8030073. [PMID: 32961854 PMCID: PMC7560251 DOI: 10.3390/toxics8030073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/31/2022]
Abstract
Water-pipe smoking (WPS) is becoming the most popular form of tobacco use among the youth, especially in the Middle East, replacing cigarettes rapidly and becoming a major risk of tobacco addiction worldwide. Smoke from WPS contains similar toxins as those present in cigarette smoke and is linked directly with different types of cancers including lung and head and neck (HN) carcinomas. However, the underlying molecular pathways and/or target genes responsible for the carcinogenic process are still unknown. In this study, human normal oral epithelial (HNOE) cells, NanoString PanCancer Pathways panel of 770 gene transcripts and quantitative real-time polymerase chain reaction (qRT-PCR) analysis were applied to discover differentially expressed genes (DEG) modulated by WPS. In silico analysis was performed to analyze the impact of these genes in HN cancer patient’s biology and outcome. We found that WPS can induce the epithelial–mesenchymal transition (EMT: hallmark of cancer progression) of HNOE cells. More significantly, our analysis of NanoString revealed 23 genes deregulated under the effect of WPS, responsible for the modulation of cell cycle, proliferation, migration/invasion, apoptosis, signal transduction, and inflammatory response. Further analysis was performed using qRT-PCR of HNOE WPS-exposed and unexposed cells supported the reliability of our NanoString data. Moreover, we demonstrate those DEG to be upregulated in cancer compared with normal tissue. Using the Kaplan–Meier analysis, we observed a significant association between WPS-deregulated genes and relapse-free survival/overall survival in HN cancer patients. Our findings imply that WPS can modulate EMT as well as a set of genes that are directly involved in human HN carcinogenesis, thereby affecting HN cancer patients’ survival.
Collapse
|
19
|
Zhu K, Li P, Mo Y, Wang J, Jiang X, Ge J, Huang W, Liu Y, Tang Y, Gong Z, Liao Q, Li X, Li G, Xiong W, Zeng Z, Yu J. Neutrophils: Accomplices in metastasis. Cancer Lett 2020; 492:11-20. [PMID: 32745581 DOI: 10.1016/j.canlet.2020.07.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/14/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
Metastasis is a critical cause of treatment failure and death in patients with advanced malignancies. Tumor cells can leave the primary site and enter the bloodstream; these circulating tumor cells then colonize target organs by overcoming blood shear stress, evading immune surveillance, and silencing the offensive capabilities of immune cells, eventually forming metastatic foci. From leaving the primary focus to the completion of distant metastasis, malignant tumor cells are supported and/or antagonized by certain immune cells. In particular, it has been found that myeloid granulocytes play an important role in this process. This review therefore aims to comprehensively describe the significance of neutrophils in solid tumor metastasis in terms of their supporting role in initiating the invasion and migration of tumor cells and assisting the colonization of circulating tumor cells in distant target organs, with the hope of providing insight into and ideas for anti-tumor metastasis treatment of tumor patients.
Collapse
Affiliation(s)
- Kunjie Zhu
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Panchun Li
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jie Wang
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Weilun Huang
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Liu
- Department of Plastic and Cosmetic Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanyan Tang
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Jianjun Yu
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Shen X, Jiang H, Chen Z, Lu B, Zhu Y, Mao J, Chai K, Chen W. MicroRNA-145 Inhibits Cell Migration and Invasion in Colorectal Cancer by Targeting TWIST. Onco Targets Ther 2019; 12:10799-10809. [PMID: 31849487 PMCID: PMC6911328 DOI: 10.2147/ott.s216147] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/31/2019] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION MicroRNAs function as oncogenes or tumor suppressors in the development of various human cancers. We investigated the effect of microRNA-145 (miR-145) on colorectal cancer (CRC) cell invasion and migration. METHODS The levels of miR-145 in CRC cells were examined by quantitative PCR; Western blotting was used to detect TWIST1 (twist family bHLH transcription factor 1) protein and the epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, vimentin). Then, we transfected miR-145 mimics or inhibitor into CRC cells and used the wound healing and Transwell invasion assays to investigate their migration and invasive capability, respectively. RESULTS The miR-145 mimics suppressed CRC cell invasion and migration significantly; in contrast, miR-145 downregulation had the opposite effect. Furthermore, miR-145 regulated TWIST1 levels negatively at transcriptional level. TWIST1 knockdown significantly inhibited the CRC cell migration ability and the number of CRC cells that crossed the Transwell membrane. There was no significant difference in terms of migration and invasive capability after the cells had been transfected with miR-145 mimics or inhibitor plus TWIST1 small interfering RNA (siRNA) as compared to the TWIST1 siRNA-only group. Furthermore, we demonstrate that the inhibition of miR-145 could enhance the capability for lung metastasis in vivo. CONCLUSION Taken together, these findings indicate that miR-145 acts as a new tumor suppressor by regulating TWIST1 and plays a vital role in the invasive and migration ability of CRC cells.
Collapse
Affiliation(s)
- Xuning Shen
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Honggang Jiang
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Zhiheng Chen
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Bohao Lu
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Yi Zhu
- Department of Gastroenterological Surgery, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Jiayan Mao
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang310012, People’s Republic of China
| | - Kequn Chai
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang310012, People’s Republic of China
- Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang310012, People’s Republic of China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang310012, People’s Republic of China
- Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang310012, People’s Republic of China
| |
Collapse
|
21
|
Li W, Cho MY, Lee S, Jang M, Park J, Park R. CRISPR-Cas9 mediated CD133 knockout inhibits colon cancer invasion through reduced epithelial-mesenchymal transition. PLoS One 2019; 14:e0220860. [PMID: 31393941 PMCID: PMC6687161 DOI: 10.1371/journal.pone.0220860] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/24/2019] [Indexed: 12/27/2022] Open
Abstract
We previously reported that CD133, as a putative cancer stem cell marker, plays an important role in cell proliferation and invasion in colon cancer. To understand the role of CD133 expression in colon cancer, we evaluated the inhibitory effect of CD133 in colon cancer cells. In this study, we generated CD133knockout colon cancer cells (LoVo) using the CRISPR-Cas9 gene editing system. CD133+ colon cancer cells (LoVo) were infected with the lentiviral vector carrying CD133 gRNA and purified cell by culturing single cell colonies. CD133knockout cells was validated by western blot and flow cytometry analysis. In functional study, we observed a significant reduction in cell proliferation and colony formation in CRISPR-Cas9 mediated CD133 knockout cells in compare with control (P < 0.001). We also found the anticancer effect of stattic was dependent on CD133 expression in colon cancer cells. Although CD133knockout cells could not completely block the tumorigenic property, they showed remarkable inhibitory effects on the ability of cell migration and invasion (P < 0.001). In addition, we examined the epithelial mesenchymal transition (EMT)-related protein expression by western blot. The result clearly showed a loss of vimentin expression in CD133knockout cells. Therefore, CRISPR-Cas9 mediated CD133knockout can be an effective treatment modality for CD133+ colon cancer through reducing the characteristics of cancer stem cells.
Collapse
Affiliation(s)
- Wanlu Li
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Mee-Yon Cho
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, South Korea
- * E-mail:
| | - Suji Lee
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Mirae Jang
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Rackhyun Park
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| |
Collapse
|
22
|
Sadek KW, Haik MY, Ashour AA, Baloch T, Aboulkassim T, Yasmeen A, Vranic S, Zeidan A, Al Moustafa AE. Water-pipe smoking promotes epithelial-mesenchymal transition and invasion of human breast cancer cells via ERK1/ERK2 pathways. Cancer Cell Int 2018; 18:180. [PMID: 30473629 PMCID: PMC6234648 DOI: 10.1186/s12935-018-0678-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022] Open
Abstract
Background With the increasing popularity of water-pipe smoking (WPS), it is critical to comprehend how WPS may affect women's health. The main goal of this study is to identify the potential outcome of WPS on human breast cancer progression. Methods Two breast cancer cell lines, MCF7 and BT20, were used in this investigation. We explored the outcome of WPS on cell morphology and cell invasion using inverted microscope and Biocoat Matrigel invasion chambers. On the other hand, Western blot was employed to study the expression patterns of key control genes of cell adhesion and invasion. Results Our data reveal that WPS induces epithelial-mesenchymal transition (EMT) of MCF7 and BT20 breast cancer cell lines; thus, WPS enhances cell invasion ability of both cell lines in comparison with their matched controls. More significantly, WPS provokes a down- and up-regulation of E-cadherin and focal adhesion kinase (FAK), respectively, which are important key regulators of cancer progression genes. Finally, our data point out that WPS incites the activation of Erk1/Erk2, which could be behind the stimulation of EMT and invasion as well as the deregulation of E-cadherin and FAK expression. Conclusion Our data show, for the first time, that WPS initiates EMT and stimulates cell invasion of breast cancer cells, which could incite metastatic development in breast cancer patients. Thus, we believe that further studies, both in vitro and in vivo, are required to elucidate the pathogenic outcome of WPS on cancer progression of several human carcinomas including breast.
Collapse
Affiliation(s)
- Khaled W Sadek
- 1College of Medicine, Qatar University, Doha, Qatar.,2Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Mahmoud Y Haik
- 1College of Medicine, Qatar University, Doha, Qatar.,2Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Anas A Ashour
- 1College of Medicine, Qatar University, Doha, Qatar.,2Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Tahira Baloch
- 3Segal Cancer Centre, Lady Davis Institute for Medical Research of the Sir Mortimer B. Davis-Jewish General Hospital, Montreal, QC Canada
| | - Tahar Aboulkassim
- 3Segal Cancer Centre, Lady Davis Institute for Medical Research of the Sir Mortimer B. Davis-Jewish General Hospital, Montreal, QC Canada
| | - Amber Yasmeen
- 3Segal Cancer Centre, Lady Davis Institute for Medical Research of the Sir Mortimer B. Davis-Jewish General Hospital, Montreal, QC Canada
| | - Semir Vranic
- 1College of Medicine, Qatar University, Doha, Qatar
| | - Asad Zeidan
- 1College of Medicine, Qatar University, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- 1College of Medicine, Qatar University, Doha, Qatar.,2Biomedical Research Centre, Qatar University, Doha, Qatar.,4Oncology Department, McGill University, Montreal, QC Canada.,Syrian Research Cancer Centre of the Syrian Society against Cancer, Aleppo, Syria
| |
Collapse
|
23
|
Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: A review. J Cell Physiol 2018; 234:5683-5699. [PMID: 30341914 DOI: 10.1002/jcp.27411] [Citation(s) in RCA: 505] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Abstract
Cyclooxygenase-2 (COX-2) is frequently expressed in many types of cancers exerting a pleiotropic and multifaceted role in genesis or promotion of carcinogenesis and cancer cell resistance to chemo- and radiotherapy. COX-2 is released by cancer-associated fibroblasts (CAFs), macrophage type 2 (M2) cells, and cancer cells to the tumor microenvironment (TME). COX-2 induces cancer stem cell (CSC)-like activity, and promotes apoptotic resistance, proliferation, angiogenesis, inflammation, invasion, and metastasis of cancer cells. COX-2 mediated hypoxia within the TME along with its positive interactions with YAP1 and antiapoptotic mediators are all in favor of cancer cell resistance to chemotherapeutic drugs. COX-2 exerts most of the functions through its metabolite prostaglandin E2. In some and limited situations, COX-2 may act as an antitumor enzyme. Multiple signals are contributed to the functions of COX-2 on cancer cells or its regulation. Members of mitogen-activated protein kinase (MAPK) family, epidermal growth factor receptor (EGFR), and nuclear factor-κβ are main upstream modulators for COX-2 in cancer cells. COX-2 also has interactions with a number of hormones within the body. Inhibition of COX-2 provides a high possibility to exert therapeutic outcomes in cancer. Administration of COX-2 inhibitors in a preoperative setting could reduce the risk of metastasis in cancer patients. COX-2 inhibition also sensitizes cancer cells to treatments like radio- and chemotherapy. Chemotherapeutic agents adversely induce COX-2 activity. Therefore, choosing an appropriate chemotherapy drugs along with adjustment of the type and does for COX-2 inhibitors based on the type of cancer would be an effective adjuvant strategy for targeting cancer.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Eniseh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
24
|
Cai J, Huang L, Huang J, Kang L, Lin H, Huang P, Zhu P, Wang J, Dong J, Wang L, Xian CJ. Associations between the cyclooxygenase-2 expression in circulating tumor cells and the clinicopathological features of patients with colorectal cancer. J Cell Biochem 2018; 120:4935-4941. [PMID: 30260024 DOI: 10.1002/jcb.27768] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/06/2018] [Indexed: 12/26/2022]
Abstract
While previous studies have shown that the number of circulating tumor cells (CTCs) alone is not sufficient to reflect tumor progression and that cyclooxygenase-2 (COX-2) expression is correlated with colorectal cancer (CRC) metastasis, COX-2 expression status and its potential functions in CTCs of CRC patients are unknown. Here, epithelial-mesenchymal transition (EMT) phenotype-based subsets of CTCs and the COX-2 expression status in CTCs were identified and their potential clinical values were assessed in 91 CRC patients. CTCs were enumerated in peripheral blood and subsets of CTCs (epithelial [eCTCs], mesenchymal [mCTCs], and biophenotypic [bCTCs]) and the COX-2 expression status were determined using the RNA in situ hybridization method. CTCs were detected in 80.2% (73 of 91) patients. Neither the total CTC nor eCTC numbers were found to significantly associate with any of the clinicopathological features. However, the number of mCTCs was significantly associated with distance metastasis (P = 0.035) and had a trend of being associated with lymph node metastasis ( P = 0.055). Among the 73 patients enrolled for evaluating COX-2 expression, 52.5% (38 of 73) were found to express COX-2 in CTCs, and COX-2 expression in CTCs was not found to associate with the clinicopathological factors. However, COX-2 expression in mCTCs tended to have a higher rate in patients with metastasis compared with those without metastasis (72.0% vs 42.8%; P = 0.072). Furthermore, COX-2 expression and mCTC marker expression correlated positively ( R = 0.287; P = 0.017). Further studies are required to investigate the clinical value of the expression of COX-2 in mCTCs, especially in CRC patients with the advanced tumor stage and distant metastasis.
Collapse
Affiliation(s)
- Jinlin Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Kang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongcheng Lin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pinzhu Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peixuan Zhu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianghui Dong
- UniSA Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Liping Wang
- UniSA Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Cory J Xian
- UniSA Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
25
|
Tong D, Liu Q, Wang LA, Xie Q, Pang J, Huang Y, Wang L, Liu G, Zhang D, Lan W, Jiang J. The roles of the COX2/PGE2/EP axis in therapeutic resistance. Cancer Metastasis Rev 2018; 37:355-368. [PMID: 30094570 DOI: 10.1007/s10555-018-9752-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|