1
|
Aksoy A, Varoglu A, Onalan EE, Tektemur A, Artas G, Koc M, Cakmak M, Aydin S, Kilic M, Ulas M. The knockdown of stathmin with si-RNA inhibits invasion of mesothelioma. Tissue Cell 2024; 87:102303. [PMID: 38244401 DOI: 10.1016/j.tice.2024.102303] [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: 06/07/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND To investigate the mechanism of action of stathmin1 (STMN1) in mesothelioma (MSM) and whether it has any role in its treatment. METHODS STMN1 expression was examined using immunohistochemistry in biopsy tissues taken from MSM patients. The relationships between the levels of STMN1 expression in the pathology preparations of MSM patients, and the clinicopathological characteristics of these patients, and their survival times were investigated. Transfection of STMN1-specific siRNA into SPC212 cells was compared to negative control siRNAs. The mRNA levels of genes that may play a role in invasion, apoptosis, and autophagy were evaluated by RT-PCR. RESULTS The expression of STMN1 was shown to be high in MSM tissues (p < 0.05). It was found that the only independent predictor factor affecting the survival time of MSM patients was the disease stage (p < 0.05). STMN1 was significantly reduced after siRNA intervention (81.5%). STMN1 with specific siRNA has been shown to suppress invasion by reducing the mRNA levels of cadherin-6 (CDH6), fibroblast growth factor-8 (FGF8), hypoxia-inducible factor 1 (HIF1A), matrix metallopeptidase 1-2 (gelatinase A) (MMP1-2), and TIMP metallopeptidase inhibitor 2 (TIMP2), which are important markers for invasion. Although the expression of apoptosis and autophagy-related genes, caspase-2 (Casp2) and LC-3, was reduced by silencing STMN1 with specific siRNA in western blot analysis, this effect was not observed in PCR results. CONCLUSIONS Immunohistochemical analysis of STMN1 may contribute to the differential diagnosis of MSM, and STMN1 may also be considered as a potential therapeutic target in the early invasive stage of MSM therapy.
Collapse
Affiliation(s)
- Asude Aksoy
- Department of Medical Oncology, University of Health Sciences, Fethi Sekin City Hospital SUAM, Elazig, Turkey.
| | - Asuman Varoglu
- Department of Neurology, Medical Faculty, Medeniyet University, Istanbul, Turkey
| | - Ebru Etem Onalan
- Department of Medical Biology and Genetics, Firat University, Elazig, Turkey
| | - Ahmet Tektemur
- Department of Medical Biology and Genetics, Firat University, Elazig, Turkey
| | - Gokhan Artas
- Department of Pathology, Medical Faculty, Firat University, Elazig, Turkey
| | - Mustafa Koc
- Department of Radiology, Medical Faculty, Firat University, Elazig, Turkey
| | - Muharrem Cakmak
- Department of Thoracic Surgery, Medical Faculty, Firat University, Elazig, Turkey
| | - Siyami Aydin
- Department of Thoracic Surgery, Medical Faculty, Firat University, Elazig, Turkey
| | - Murat Kilic
- Department of Thoracic Surgery, Inonu University, Malatya, Turkey
| | - Mustafa Ulas
- Department of Physiology, Medical Faculty, Firat University, Elazig, Turkey
| |
Collapse
|
2
|
Chang CC, Takada YK, Cheng CW, Maekawa Y, Mori S, Takada Y. FGF9, a Potent Mitogen, Is a New Ligand for Integrin αvβ3, and the FGF9 Mutant Defective in Integrin Binding Acts as an Antagonist. Cells 2024; 13:307. [PMID: 38391921 PMCID: PMC10887216 DOI: 10.3390/cells13040307] [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: 12/28/2023] [Revised: 01/24/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
FGF9 is a potent mitogen and survival factor, but FGF9 protein levels are generally low and restricted to a few adult organs. Aberrant expression of FGF9 usually results in cancer. However, the mechanism of FGF9 action has not been fully established. Previous studies showed that FGF1 and FGF2 directly bind to integrin αvβ3, and this interaction is critical for signaling functions (FGF-integrin crosstalk). FGF1 and FGF2 mutants defective in integrin binding were defective in signaling, whereas the mutants still bound to FGFR suppressed angiogenesis and tumor growth, indicating that they act as antagonists. We hypothesize that FGF9 requires direct integrin binding for signaling. Here, we show that docking simulation of the interaction between FGF9 and αvβ3 predicted that FGF9 binds to the classical ligand-binding site of αvβ3. We show that FGF9 bound to integrin αvβ3 and generated FGF9 mutants in the predicted integrin-binding interface. An FGF9 mutant (R108E) was defective in integrin binding, activating FRS2α and ERK1/2, inducing DNA synthesis, cancer cell migration, and invasion in vitro. R108E suppressed DNA synthesis and activation of FRS2α and ERK1/2 induced by WT FGF9 (dominant-negative effect). These findings indicate that FGF9 requires direct integrin binding for signaling and that R108E has potential as an antagonist to FGF9 signaling.
Collapse
Affiliation(s)
- Chih-Chieh Chang
- Department of Dermatology, University of California, Davis School of Medicine, Sacramento, CA 95817, USA; (C.-C.C.); (Y.K.T.)
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Sacramento, CA 95817, USA
| | - Yoko K. Takada
- Department of Dermatology, University of California, Davis School of Medicine, Sacramento, CA 95817, USA; (C.-C.C.); (Y.K.T.)
| | - Chao-Wen Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yukina Maekawa
- Department of Medical Technology, Faculty of Health Science, Morinomiya University of Medical Sciences, Osaka 536-0025, Japan; (Y.M.); (S.M.)
| | - Seiji Mori
- Department of Medical Technology, Faculty of Health Science, Morinomiya University of Medical Sciences, Osaka 536-0025, Japan; (Y.M.); (S.M.)
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yoshikazu Takada
- Department of Dermatology, University of California, Davis School of Medicine, Sacramento, CA 95817, USA; (C.-C.C.); (Y.K.T.)
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
3
|
Chang CC, Takada YK, Cheng CW, Maekawa Y, Mori S, Takada Y. FGF9, a potent mitogen, is a new ligand for integrin αvβ3, and the FGF9 mutant defective in integrin binding acts as an antagonist. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569657. [PMID: 38076804 PMCID: PMC10705552 DOI: 10.1101/2023.12.01.569657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
FGF9 is a potent mitogen and survival factor, but FGF9 protein level is generally low and restricted to a few adult organs. Aberrant expression of FGF9 usually results in cancer. However, the mechanism of FGF9 action has not been fully established. Previous studies showed that FGF1 and FGF2 directly bind to integrin αvβ3 and this interaction is critical for signaling functions (FGF-integrin crosstalk). FGF1 and FGF2 mutants defective in integrin binding were defective in signaling, whereas the mutants still bound to FGFR, and suppressed angiogenesis and tumor growth, indicating that they act as antagonists. We hypothesize that FGF9 requires direct integrin binding for signaling. Here we show that docking simulation of interaction between FGF9 and αvβ3 predicted that FGF9 binds to the classical ligand-binding site of αvβ3. We showed that FGF9 actually bound to integrin αvβ3, and generated an FGF9 mutants in the predicted integrin-binding interface. An FGF9 mutant (R108E) was defective in integrin binding, activating FRS2α and ERK1/2, inducing DNA synthesis, cancer cell migration, and invasion in vitro. R108E suppressed DNA synthesis induced by WT FGF9 and suppressed DNA synthesis and activation of FRS2α and ERK1/2 induced by WT FGF9 (dominant-negative effect). These findings indicate that FGF9 requires direct integrin binding for signaling and that R108E has potential as an antagonist to FGF9 signaling.
Collapse
|
4
|
FGF9 promotes cell proliferation and tumorigenesis in TM3 mouse Leydig progenitor cells. Am J Cancer Res 2022; 12:5613-5630. [PMID: 36628285 PMCID: PMC9827084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/06/2022] [Indexed: 01/12/2023] Open
Abstract
Fibroblast growth factor 9 (FGF9) modulates cell proliferation, differentiation and motility for development and tissue repair in normal cells. Growing evidence shows that abnormal activation of FGF9 signaling is associated with tumor malignancy. We have previously reported that FGF9 increases MA-10 mouse Leydig tumor cell proliferation, in vitro, and tumor growth, in vivo. Also, FGF9 promotes the tumor growth and liver metastasis of mouse Lewis lung cancer cells, in vivo. However, the effects of FGF9 in the early stage of tumorigenesis remains elusive. In this study, TM3 mouse Leydig progenitor cells, that are not tumorigenic in immunocompromised mice, were used as a model cell line to investigate the role of FGF9 in tumorigenesis. The results demonstrated that FGF9 significantly induced cell proliferation and activated the MAPK, PI3K and PLCγ signaling pathways in TM3 cells. The percentage of the cell number in G1 phase was reduced and that in S and G2/M phases was increased after FGF9 stimulation in TM3 cells. Cyclin D1, cyclin A1, CDK2, CDK1, and p21 expressions and the phosphorylation level of Rb were all induced in FGF9-treated TM3 cells. In addition, FGF9 increased the expression of FGF receptor 1-4 in TM3 cells, suggesting the positive feedback loop between FGF9 and FGFRs. Furthermore, in the allograft mouse model, FGF9 promoted the tumorigenesis of TM3 cells characterized by higher expression of tumor markers, such as tumor necrosis factor alpha (TNFα) and α-fetoprotein (AFP), in the subcutaneously inoculated TM3 cell tissue. Conclusively, FGF9 induced cell cycle to increase cell proliferation of TM3 cells through FAK, MAPK, PI3K/Akt and PLCγ signaling pathways, in vitro, and promoted the tumorigenesis of TM3 cell allograft tissue, in vivo, which is a potential marker for tumor as well as a target for cancer therapeutic strategies.
Collapse
|
5
|
Zeng S, Feng X, Xing S, Xu Z, Miao Z, Liu Q. Advanced Peptide Nanomedicines for Bladder Cancer Theranostics. Front Chem 2022; 10:946865. [PMID: 35991612 PMCID: PMC9389364 DOI: 10.3389/fchem.2022.946865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is still a global public health problem. Although remarkable success has been achieved in cancer diagnosis and treatment, the high recurrence and mortality rates remain severely threatening to human lives and health. In recent years, peptide nanomedicines with precise selectivity and high biocompatibility have attracted intense attention in biomedical applications. In particular, there has been a significant increase in the exploration of peptides and their derivatives for malignant tumor therapy and diagnosis. Herein, we review the applications of peptides and their derivatives in the diagnosis and treatment of bladder cancer, providing new insights for the design and development of novel peptide nanomedicines for the treatment of bladder cancer in the future.
Collapse
Affiliation(s)
- Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| | - Xiaodi Feng
- Department of Urology, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), ShanDong, China
| | - Shaoqiang Xing
- Department of Urology, Weihai Central Hospital, ShanDong, China
| | - Zhaoliang Xu
- Department of Urology, First Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Zhizhao Miao
- School of Medicine, Nankai University, Tianjin, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
- *Correspondence: Qian Liu,
| |
Collapse
|
6
|
Chang MM, Wu SZ, Yang SH, Wu CC, Wang CY, Huang BM. FGF9/FGFR1 promotes cell proliferation, epithelial-mesenchymal transition, M2 macrophage infiltration and liver metastasis of lung cancer. Transl Oncol 2021; 14:101208. [PMID: 34438248 PMCID: PMC8390529 DOI: 10.1016/j.tranon.2021.101208] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/18/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022] Open
Abstract
FGF9 induced cell proliferation, EMT, migration, and invasion of mouse Lewis lung cancer (LLC) cells, in vitro. FGF9 interacted with FGFR1 and activated FAK, AKT, and ERK/MAPK signal pathways, induced the expression of EMT key proteins (N-cadherin, vimentin, snail, MMP2, MMP3 and MMP13) and reduced the expression of E-cadherin. FGF9 promoted liver metastasis of subcutaneous inoculated LLC tumor with tumor growth, angiogenesis, EMT and M2-macrophage infiltration in the tumor microenvironment. The FGF9/LLC syngeneic animal model provides a useful tool for the mechanism studies of liver metastasis which is the worst prognostic factor for lung cancer patients with distant organ metastasis.
Fibroblast growth factors 9 (FGF9) modulates cell proliferation, differentiation and motility for development and repair in normal cells. Abnormal activation of FGF9 signaling is associated with tumor progression in many cancers. Also, FGF9 may be an unfavorable prognostic indicator for non-small cell lung cancer patients. However, the effects and mechanisms of FGF9 in lung cancer remain elusive. In this study, we investigated the FGF9-induced effects and signal activation profiles in mouse Lewis lung carcinoma (LLC) in vitro and in vivo. Our results demonstrated that FGF9 significantly induced cell proliferation and epithelial-to-mesenchymal transition (EMT) phenomena (migration and invasion) in LLC cells. Mechanism-wise, FGF9 interacted with FGFR1 and activated FAK, AKT, and ERK/MAPK signal pathways, induced the expression of EMT key proteins (N-cadherin, vimentin, snail, MMP2, MMP3 and MMP13), and reduced the expression of E-cadherin. Moreover, in the allograft mouse model, intratumor injection of FGF9 to LLC-tumor bearing C57BL/6 mice enhanced LLC tumor growth which were the results of increased Ki67 expression and decreased cleaved caspase-3 expression compared to control groups. Furthermore, we have a novel finding that FGF9 promoted liver metastasis of subcutaneous inoculated LLC tumor with angiogenesis, EMT and M2-macrophage infiltration in the tumor microenvironment. In conclusion, FGF9 activated FAK, AKT, and ERK signaling through FGFR1 with induction of EMT to stimulate LLC tumorigenesis and hepatic metastasis. This novel FGF9/LLC allograft animal model may therefore be useful to study the mechanism of liver metastasis which is the worst prognostic factor for lung cancer patients with distant organ metastasis.
Collapse
Affiliation(s)
- Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, Republic of China
| | - Su-Zhen Wu
- Department of Anesthesiology, Chi Mei Medical Center, Liouying, Tainan 73657, Taiwan, Republic of China
| | - Shang-Hsun Yang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, Republic of China
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, Republic of China
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, Republic of China.
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, Republic of China; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40406, Taiwan, Republic of China.
| |
Collapse
|
7
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
8
|
Habibi H, Suzuki A, Hayashi K, Salimi H, Terai H, Hori Y, Tamai K, Orita K, Ohyama S, Yabu A, Maruf MH, Nakamura H. Expression and function of FGF9 in the hypertrophied ligamentum flavum of lumbar spinal stenosis patients. Spine J 2021; 21:1010-1020. [PMID: 33577925 DOI: 10.1016/j.spinee.2021.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/23/2020] [Accepted: 02/06/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Ligamentum flavum (LF) hypertrophy plays a dominant role in lumbar spinal stenosis (LSS). A previous study found that fibroblast growth factor 9 (FGF9) was upregulated with mechanical stress in rabbit LF. However, the expression and function of FGF9 are not well understood in human LF. PURPOSE To evaluate FGF9 expression and function in human LF with and without hypertrophy. STUDY DESIGN This study employed a basic research study design utilizing human LF tissue for histological analyses. PATIENT SAMPLES Hypertrophied LF tissue sample from patients with LSS, and nonhypertrophied (control) LFs from patients with lumbar disc herniation or other diseases were obtained during surgery. METHODS LF specimens were histologically analyzed for FGF9 and vascular endothelial growth factor A (VEGF-A) by immunohistochemistry. The number of total and FGF9 immuno-positive cells and blood vessels were counted and compared between LF with and without hypertrophy. For functional analysis, the effect of FGF9 on cell proliferation and migration was examined using a primary cell culture of human LF. RESULTS Histological studies revealed that the total cell number was significantly higher in the LF of patients with LSS than in the LF of control patients. Immunohistochemistry showed that the percentage of FGF9-positive cells was significantly higher in the LF of patients with LSS than in the controls, and it positively correlated with patients' age, regardless of disease. Double immune-positive cells for FGF9 and VEGF-A were often observed in vascular endothelial cells and fibroblasts in the fibrotic area of hypertrophied LF, and the number of double positive vessels was significantly higher in LF of LSS patients than in the LF of controls. Primary cell culture of human LF revealed that FGF9 promoted the proliferation and migration of LF cells. CONCLUSION The present study demonstrated that FGF9 expression is highly upregulated in hypertrophied human LF. FGF9 potentially plays a pivotal role in the process of hypertrophy of LF, which is associated with mechanical stress, through cell proliferation and migration. CLINICAL SIGNIFICANCE The results from this study partially reveal the molecular mechanisms of LF hypertrophy and suggest that FGF9 may be involved in the process of LF degeneration in elderly patients.
Collapse
Affiliation(s)
- Hasibullah Habibi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akinobu Suzuki
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - Kazunori Hayashi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hamidullah Salimi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hidetomi Terai
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Hori
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Tamai
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kumi Orita
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shoichiro Ohyama
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akito Yabu
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Mohammad Hasib Maruf
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
9
|
Chan LC, Kalyanasundram J, Leong SW, Masarudin MJ, Veerakumarasivam A, Yusoff K, Chan SC, Chia SL. Persistent Newcastle disease virus infection in bladder cancer cells is associated with putative pro-survival and anti-viral transcriptomic changes. BMC Cancer 2021; 21:625. [PMID: 34044804 PMCID: PMC8161962 DOI: 10.1186/s12885-021-08345-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 05/12/2021] [Indexed: 12/15/2022] Open
Abstract
Background Newcastle disease virus (NDV) is an oncolytic virus with excellent selectivity against cancer cells, both in vitro and in vivo. Unfortunately, prolonged in vitro NDV infection results in the development of persistent infection in the cancer cells which are then able to resist NDV-mediated oncolysis. However, the mechanism of persistency of infection remains poorly understood. Methods In this study, we established persistently NDV-infected EJ28 bladder cancer cells, designated as EJ28P. Global transcriptomic analysis was subsequently carried out by microarray analysis. Differentially expressed genes (DEGs) between EJ28 and EJ28P cells identified by the edgeR program were further analysed by Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) analyses. In addition, the microarray data were validated by RT-qPCR. Results Persistently NDV-infected EJ28 bladder cancer cells were successfully established and confirmed by flow cytometry. Microarray analysis identified a total of 368 genes as differentially expressed in EJ28P cells when compared to the non-infected EJ28 cells. GSEA revealed that the Wnt/β-catenin and KRAS signalling pathways were upregulated while the TGF-β signalling pathway was downregulated. Findings from this study suggest that the upregulation of genes that are associated with cell growth, pro-survival, and anti-apoptosis may explain the survivability of EJ28P cells and the development of persistent infection of NDV. Conclusions This study provides insights into the transcriptomic changes that occur and the specific signalling pathways that are potentially involved in the development and maintenance of NDV persistency of infection in bladder cancer cells. These findings warrant further investigation and is crucial towards the development of effective NDV oncolytic therapy against cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08345-y.
Collapse
Affiliation(s)
- Lee-Chin Chan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor Darul Ehsan, Malaysia.,Malaysia Genome Institute, Ministry of Science, Technology and Innovation, Jalan Bangi, 43000, Kajang, Selangor Darul Ehsan, Malaysia
| | - Jeevanathan Kalyanasundram
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Sze-Wei Leong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Mas Jaffri Masarudin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| | - Abhi Veerakumarasivam
- Malaysia Genome Institute, Ministry of Science, Technology and Innovation, Jalan Bangi, 43000, Kajang, Selangor Darul Ehsan, Malaysia.,Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.,Medical Genetics Laboratory, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor Darul Ehsan, Malaysia.,Malaysia Genome Institute, Ministry of Science, Technology and Innovation, Jalan Bangi, 43000, Kajang, Selangor Darul Ehsan, Malaysia.,UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| | - Soon-Choy Chan
- Malaysia Genome Institute, Ministry of Science, Technology and Innovation, Jalan Bangi, 43000, Kajang, Selangor Darul Ehsan, Malaysia. .,Perdana University School of Liberal Arts, Science and Technology (PUScLST), Perdana University, 50490, Kuala Lumpur, Malaysia.
| | - Suet-Lin Chia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor Darul Ehsan, Malaysia. .,UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
10
|
Miki Y, Yashiro M, Moyano-Galceran L, Sugimoto A, Ohira M, Lehti K. Crosstalk Between Cancer Associated Fibroblasts and Cancer Cells in Scirrhous Type Gastric Cancer. Front Oncol 2020; 10:568557. [PMID: 33178597 PMCID: PMC7596590 DOI: 10.3389/fonc.2020.568557] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause among all cancer deaths globally. Although the treatment outcome of GC has improved, the survival of patients with GC at stages III and IV remains unsatisfactory. Among several types of GC, scirrhous type GC (SGC) shows highly aggressive growth and invasive activity, leading to frequent peritoneal metastasis. SGC is well known to accompany abundant stromal cells that compose the tumor microenvironment (TME) along with the produced extracellular matrix (ECM) and secreted factors. One of the main stromal components is cancer associated fibroblast (CAF). In the SGC microenvironment, CAFs are a source of various secreted factors, including fibroblast growth factors (FGFs), which mediate prominent tumor-stimulating activity. In turn, cancer cells also secrete numerous factors, which can activate and educate CAFs. Current findings suggest that cancer cells and stromal cells communicate interactively via the soluble factors, the ECM, and likely also by exosomes. In this review, we focus on the soluble factors mediating communication between cancer cells and CAFs in SGC, and consider how they are related to the modulation of TME and the high rate of peritoneal metastasis. At last, we discuss the perspectives on targeting these communication pathways for improved future treatment.
Collapse
Affiliation(s)
- Yuichiro Miki
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Masakazu Yashiro
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Atsushi Sugimoto
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
11
|
Paur J, Valler M, Sienel R, Taxauer K, Holzmann K, Marian B, Unterberger A, Mohr T, Berger W, Gvozdenovich A, Schimming J, Grusch M, Grasl‐Kraupp B. Interaction of FGF9 with FGFR3-IIIb/IIIc, a putative driver of growth and aggressive behaviour of hepatocellular carcinoma. Liver Int 2020; 40:2279-2290. [PMID: 32378800 PMCID: PMC7496895 DOI: 10.1111/liv.14505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/15/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Recently, overexpression of the fibroblast growth factor receptor 3 (FGFR3) splice variants FGFR3-IIIb and FGFR3-IIIc was found in ~50% of hepatocellular carcinoma (HCC). Here, we aim to identify FGFR3-IIIb/IIIc ligands, which drive the progression of HCC. METHODS FACS, MTT assay and/or growth curves served to identify the FGFR3-IIIb/IIIc ligand being most effective to induce growth of hepatoma/hepatocarcinoma cell lines, established from human HCC. The most potent FGF was characterized regarding the expression levels in epithelial and stromal cells of liver and HCC and impact on neoangiogenesis, clonogenicity and invasive growth of hepatoma/hepatocarcinoma cells. RESULTS Among all FGFR3-IIIb/IIIc ligands tested, FGF9 was the most potent growth factor for hepatoma/hepatocarcinoma cells. Replication and/or sprouting of blood/lymphendothelial cells was stimulated as well. FGF9 occurred mainly in stromal cells of unaltered liver but in epithelial cells of HCC. Every fifth HCC exhibited overexpressed FGF9 and frequent co-upregulation of FGFR3-IIIb/IIIc. In hepatoma/hepatocarcinoma cells FGF9 enhanced the capability for clonogenicity and disintegration of the blood and lymphatic endothelium, being most pronounced in cells overexpressing FGFR3-IIIb or FGFR3-IIIc, respectively. Any of the FGF9 effects in hepatoma/hepatocarcinoma cells was blocked completely by applying the FGFR1-3-specific tyrosine kinase inhibitor BGJ398 or siFGFR3, while siFGFR1/2/4 were mostly ineffective. CONCLUSIONS FGF9 acts via FGFR3-IIIb/IIIc to enhance growth and aggressiveness of HCC cells. Accordingly, blockade of the FGF9-FGFR3-IIIb/IIIc axis may be an efficient therapeutic option for HCC patients.
Collapse
Affiliation(s)
- Jakob Paur
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Maximilian Valler
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Rebecca Sienel
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Karin Taxauer
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Klaus Holzmann
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Brigitte Marian
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Andreas Unterberger
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Thomas Mohr
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Walter Berger
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Andja Gvozdenovich
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Johannes Schimming
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Michael Grusch
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Bettina Grasl‐Kraupp
- Department of Medicine IDivision: Institute of Cancer ResearchComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| |
Collapse
|
12
|
Asakawa M, Itoh M, Suganami T, Sakai T, Kanai S, Shirakawa I, Yuan X, Hatayama T, Shimada S, Akiyama Y, Fujiu K, Inagaki Y, Manabe I, Yamaoka S, Yamada T, Tanaka S, Ogawa Y. Upregulation of cancer-associated gene expression in activated fibroblasts in a mouse model of non-alcoholic steatohepatitis. Sci Rep 2019; 9:19601. [PMID: 31862949 PMCID: PMC6925281 DOI: 10.1038/s41598-019-56039-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/03/2019] [Indexed: 12/24/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH), characterized by chronic inflammation and fibrosis, is predicted to be the leading cause of cirrhosis and hepatocellular carcinoma (HCC) in the next decade. Although recent evidence suggests the importance of fibrosis as the strongest determinant of HCC development, the molecular mechanisms underlying NASH-induced carcinogenesis still remain unclear. Here we performed RNA sequencing analysis to compare gene expression profiles of activated fibroblasts prepared from two distinct liver fibrosis models: carbon tetrachloride–induced fibrosis as a model without obesity and HCC and genetically obese melanocortin 4 receptor–deficient (MC4R-KO) mice fed Western diet, which develop steatosis, NASH, and eventually HCC. Our data showed that activated fibroblasts exhibited distinct gene expression patterns in each etiology, and that the ‘pathways in cancer’ were selectively upregulated in the activated fibroblasts from MC4R-KO mice. The most upregulated gene in these pathways was fibroblast growth factor 9 (FGF9), which was induced by metabolic stress such as palmitate. FGF9 exerted anti-apoptotic and pro-migratory effects in fibroblasts and hepatoma cells in vitro and accelerated tumor growth in a subcutaneous xenograft model. This study reveals upregulation of cancer-associated gene expression in activated fibroblasts in NASH, which would contribute to the progression from NASH to HCC.
Collapse
Affiliation(s)
- Masahiro Asakawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michiko Itoh
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. .,Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan. .,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan. .,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Takeru Sakai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayaka Kanai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ibuki Shirakawa
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Xunmei Yuan
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomi Hatayama
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuhito Fujiu
- Department of Advanced Cardiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan. .,Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. .,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. .,Japan Agency for Medical Research and Development, CREST, Tokyo, Japan.
| |
Collapse
|
13
|
Wang J, Tan X, Guo Q, Lin X, Huang Y, Chen L, Zeng X, Li R, Wang H, Wu X. FGF9 inhibition by a novel binding peptide has efficacy in gastric and bladder cancer per se and reverses resistance to cisplatin. Pharmacol Res 2019; 152:104575. [PMID: 31805343 DOI: 10.1016/j.phrs.2019.104575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
Abstract
Aberrant over-expressions of FGF9 in gastric cancer (GC) and its high-affinity receptor FGFR3c in bladder cancer (BC) provide possibilities for the treatment of GC and BC via targeting FGF9. In this study, we isolated a novel FGF9-binding peptide (P4) by screening a phage display random heptapeptide library. Sequence comparison showed that P4 shared high homology with the conserved motif in the immunoglobulin-like (Ig-like) domain II∼III (D2-D3) linker of the FGF9 high-affinity receptor (FGFR3c). The interaction between P4 and FGF9 was confirmed by the surface plasmon resonance (SPR) assay. Functional analysis indicated that P4 counteracted FGF9-induced aggressive phenotype, including cell proliferation, migration, and invasion in vitro, as well as suppressed tumor growth in vivovia down-regulation of the MAPKs and Akt cascades. More importantly, we found that FGF9 served as an underlying mechanism of the chemoresistance in GC and BC cells, and P4 could increase the sensitivity to the chemical agent via antagonizing the suppression effects of FGF9 on cell apoptosis. Taken together, our study identified a novel binding peptide for FGF9, which may serve as a potential therapeutic agent for malignant tumors featured by abnormally up-regulation of FGF9.
Collapse
Affiliation(s)
- Jizhong Wang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Xiangpeng Tan
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China; Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuxiao Guo
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Xiaomian Lin
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Yishan Huang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Liankuai Chen
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Xiangfeng Zeng
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Rongzhen Li
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Heng Wang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | - Xiaoping Wu
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
14
|
Zhang J, Tang PMK, Zhou Y, Cheng ASL, Yu J, Kang W, To KF. Targeting the Oncogenic FGF-FGFR Axis in Gastric Carcinogenesis. Cells 2019; 8:cells8060637. [PMID: 31242658 PMCID: PMC6627225 DOI: 10.3390/cells8060637] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most wide-spread malignancies in the world. The oncogenic role of signaling of fibroblast growing factors (FGFs) and their receptors (FGFRs) in gastric tumorigenesis has been gradually elucidated by recent studies. The expression pattern and clinical correlations of FGF and FGFR family members have been comprehensively delineated. Among them, FGF18 and FGFR2 demonstrate the most prominent driving role in gastric tumorigenesis with gene amplification or somatic mutations and serve as prognostic biomarkers. FGF-FGFR promotes tumor progression by crosstalking with multiple oncogenic pathways and this provides a rational therapeutic strategy by co-targeting the crosstalks to achieve synergistic effects. In this review, we comprehensively summarize the pathogenic mechanisms of FGF-FGFR signaling in gastric adenocarcinoma together with the current targeted strategies in aberrant FGF-FGFR activated GC cases.
Collapse
Affiliation(s)
- Jinglin Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| | - Patrick M K Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
15
|
Sawaki K, Kanda M, Kodera Y. Review of recent efforts to discover biomarkers for early detection, monitoring, prognosis, and prediction of treatment responses of patients with gastric cancer. Expert Rev Gastroenterol Hepatol 2018; 12:657-670. [PMID: 29902383 DOI: 10.1080/17474124.2018.1489233] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gastric cancer (GC) is the leading cause of cancer-related death worldwide. Despite recent advances in diagnosis and therapy, the prognosis of patients with GC is poor. Many patients have inoperable disease upon diagnosis or experience recurrent disease after curative gastrectomy. Unfortunately, tumor markers for GC, such as serum carcinoembryonic antigen and carbohydrate antigen 19-9, lack sufficient sensitivity and specificity. Therefore, effective biomarkers are required to detect early GC and to predict tumor recurrence and chemosensitivity. Areas covered: Here we aimed to review recent developments in techniques that improve the detection of aberrant expression of GC-associated molecules, including protein coding genes, microRNAs, long noncoding RNAs, and methylated promoter DNAs. Expert commentary: Detection of genetic and epigenetic alterations in gastric tissue or in the circulation will likely improve the diagnosis and management of GC to achieve significantly improved outcomes.
Collapse
Affiliation(s)
- Koichi Sawaki
- a Department of Gastroenterological Surgery (Surgery II) , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Mitsuro Kanda
- a Department of Gastroenterological Surgery (Surgery II) , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Yasuhiro Kodera
- a Department of Gastroenterological Surgery (Surgery II) , Nagoya University Graduate School of Medicine , Nagoya , Japan
| |
Collapse
|
16
|
Werner TA, Forster CM, Dizdar L, Verde PE, Raba K, Schott M, Knoefel WT, Krieg A. CXCR4/CXCR7/CXCL12 axis promotes an invasive phenotype in medullary thyroid carcinoma. Br J Cancer 2017; 117:1837-1845. [PMID: 29112684 PMCID: PMC5729476 DOI: 10.1038/bjc.2017.364] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023] Open
Abstract
Background: Medullary thyroid carcinoma (MTC) is a rare and challenging endocrine malignancy. Once spread, the therapeutic options are limited and the outcome poor. For these patients, the identification of new druggable biological markers is of great importance. Here, we investigated the prognostic and biological role of the C-X-C chemokine receptors type 4 and 7 (CXCR4/7) in MTC. Methods: Eighty-six MTC and corresponding non-neoplastic thyroid specimens were immunohistochemically stained for CXCR4/7 using tissue microarray technology and expression levels correlated with clinicopathological variables. Medullary thyroid carcinoma cell line TT was treated with recombinant human SDF1α/CXCL12 (rh-SDF1α) and CXCR4 antagonists AMD3100 and WZ811. Changes in cell cycle activation, tumour cell invasiveness as well as changes in mRNA expression levels of genes associated with epithelial–mesenchymal transition (EMT) were investigated. Results: High CXCR4 expression was associated with large tumour size and metastatic disease. CXCR4 antagonists significantly reduced tumour cell invasiveness, while the treatment with rh-SDF1α stimulated invasive growth, caused cell cycle activation and induced EMT. Conclusions: The CXCR4/CXCR7/CXCL12 axis plays an important role in MTC. We provide first evidence that the chemokine receptors might serve as potential therapeutic targets in patients with advanced MTC and offer new valuable insight into the underlying molecular machinery of metastatic MTC.
Collapse
Affiliation(s)
- Thomas A Werner
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| | - Christina M Forster
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| | - Levent Dizdar
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| | - Pablo E Verde
- Coordination Centre for Clinical Trials, Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| | - Katharina Raba
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf, 40225, Germany
| | - Matthias Schott
- Division for Specific Endocrinology, Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| | - Wolfram T Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| | - Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Duesseldorf 40225, Germany
| |
Collapse
|
17
|
Justet A, Joannes A, Besnard V, Marchal-Sommé J, Jaillet M, Bonniaud P, Sallenave JM, Solhonne B, Castier Y, Mordant P, Mal H, Cazes A, Borie R, Mailleux AA, Crestani B. FGF9 prevents pleural fibrosis induced by intrapleural adenovirus injection in mice. Am J Physiol Lung Cell Mol Physiol 2017; 313:L781-L795. [PMID: 28729349 DOI: 10.1152/ajplung.00508.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor 9 (FGF9) is necessary for fetal lung development and is expressed by epithelium and mesothelium. We evaluated the role of FGF9 overexpression on adenoviral-induced pleural injury in vivo and determined the biological effects of FGF9 on mesothelial cells in vitro. We assessed the expression of FGF9 and FGF receptors by mesothelial cells in both human and mouse lungs. Intrapleural injection of an adenovirus expressing human FGF9 (AdFGF9) or a control adenovirus (AdCont) was performed. Mice were euthanized at days 3, 5, and 14 Expression of FGF9 and markers of inflammation and myofibroblastic differentiation was studied by qPCR and immunohistochemistry. In vitro, rat mesothelial cells were stimulated with FGF9 (20 ng/ml), and we assessed its effect on proliferation, survival, migration, and differentiation. FGF9 was expressed by mesothelial cells in human idiopathic pulmonary fibrosis. FGF receptors, mainly FGFR3, were expressed by mesothelial cells in vivo in humans and mice. AdCont instillation induced diffuse pleural thickening appearing at day 5, maximal at day 14 The altered pleura cells strongly expressed α-smooth muscle actin and collagen. AdFGF9 injection induced maximal FGF9 expression at day 5 that lasted until day 14 FGF9 overexpression prevented pleural thickening, collagen and fibronectin accumulation, and myofibroblastic differentiation of mesothelial cells. In vitro, FGF9 decreased mesothelial cell migration and inhibited the differentiating effect of transforming growth factor-β1. We conclude that FGF9 has a potential antifibrotic effect on mesothelial cells.
Collapse
Affiliation(s)
- Aurélien Justet
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, Paris, France
| | - Audrey Joannes
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Valérie Besnard
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Joëlle Marchal-Sommé
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Madeleine Jaillet
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Philipe Bonniaud
- Institut National de la Santé et de la Recherche Médicale U866, Université de Bourgogne, Dijon, France
| | - Jean Michel Sallenave
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Brigitte Solhonne
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Yves Castier
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Chirurgie Thoracique et Vasculaire, Paris, France
| | - Pierre Mordant
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Chirurgie Thoracique et Vasculaire, Paris, France
| | - Hervé Mal
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie et Transplantation, Paris, France; and
| | - Aurélie Cazes
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Département d'Anatomie Pathologique, Paris, France
| | - Raphael Borie
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, Paris, France
| | - Arnaud A Mailleux
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France.,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Bruno Crestani
- Institut National de la Santé et de la Recherche Médicale U1152, Paris, France; .,Département Hospitalo-Universitaire Fibrosis Inflammation and Remodeling (DHU FIRE), Paris, France.,Labex Inflamex, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, Paris, France
| |
Collapse
|
18
|
Sun DP, Fang CL, Chen HK, Wen KS, Hseu YC, Hung ST, Uen YH, Lin KY. EPAC1 overexpression is a prognostic marker and its inhibition shows promising therapeutic potential for gastric cancer. Oncol Rep 2017; 37:1953-1960. [PMID: 28260059 PMCID: PMC5367365 DOI: 10.3892/or.2017.5442] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/01/2017] [Indexed: 01/18/2023] Open
Abstract
cAMP signaling controls a variety of cellular functions. In addition to the well-known signal transducer cAMP-dependent protein kinase, a more recently discovered transducer is the exchange protein directly activated by cAMP (EPAC). EPAC responses are mediated by small G proteins, which regulate biologic functions such as cell adhesion, migration and proliferation. Recently, the clinical importance of EPAC1 has received increased attention. This study investigated the correlations between the expression of EPAC1 and various clinicopathologic parameters as well as the survival of the patients with gastric cancer (GC). The patient cohort in this study consisted of 141 cases of GC that presented from 1999 through 2011; documented clinicopathologic parameters and clinical outcomes were available for all cases. Immunoblotting, immunohistochemistry and quantitative real-time PCR were used to examine EPAC1 expression in gastric cells and tissues. siRNA technology was used to study the effect of EPAC1 knockdown on cell proliferation and invasion. An increase in EPAC1 expression was found in GC cells and tissues. The overexpression of EPAC1 was associated with the depth of invasion (P=0.0021), stage (P=0.0429), and vascular invasion (P=0.0049) and was correlated with poor disease-free survival (P=0.0029) and overall survival (P=0.0024). A univariate Cox regression analysis showed that the overexpression of EPAC1 was a prognostic marker for GC (P=0.038). Furthermore, cell studies indicated that the knockdown of EPAC1 in GC cells suppressed cell proliferation and invasion. The overexpression of EPAC1 can be used as a marker to predict the outcome of patients with GC, and EPAC1 represents a potential therapeutic modality for treating GC.
Collapse
Affiliation(s)
- Ding-Ping Sun
- Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Chia-Lang Fang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Han-Kun Chen
- Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Kuo-Shan Wen
- Department of Pharmacy, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - You-Cheng Hseu
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan, R.O.C
| | - Shih-Ting Hung
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Yih-Huei Uen
- The Superintendent's Office, Chi Mei Hospital Chiali, Tainan, Taiwan, R.O.C
| | - Kai-Yuan Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| |
Collapse
|
19
|
Eizuka M, Sugai T, Habano W, Uesugi N, Takahashi Y, Kawasaki K, Yamamoto E, Suzuki H, Matsumoto T. Molecular alterations in colorectal adenomas and intramucosal adenocarcinomas defined by high-density single-nucleotide polymorphism arrays. J Gastroenterol 2017; 52:1158-1168. [PMID: 28197804 PMCID: PMC5666076 DOI: 10.1007/s00535-017-1317-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/30/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND We examined colorectal adenomas and intramucosal adenocarcinomas (IMAs) to develop a genome-wide overview of copy number alterations (CNAs) during colorectal tumorigenesis. METHODS We analysed CNAs using a high-resolution SNP array of isolated tumour glands obtained from 55 colorectal adenomas (35 low-grade adenomas and 20 high-grade adenomas) and 30 IMAs. Next, we examined whether frequent CNAs differed between low-grade and high-grade adenomas or high-grade adenomas and IMAs. Finally, we investigated the total lengths of the CNAs in low-grade adenomas, high-grade adenomas, and IMAs. RESULTS Although no frequent CNAs were found in low-grade adenomas, the most frequent alterations of high-grade adenomas were gains of 7q11, 7q21 and 9p13 and loss of 5q14.3-35. High levels of gains were detected at 13q, 7q, 8p, 20q, 7p, 18p and 17p in IMAs. Although no frequent alteration differed between low-grade and high-grade adenomas, significant differences of gains at 13q, 17p and 18p were found between high-grade adenoma and IMAs. Although the total lengths of all CNAs (gains and losses), copy number gains, and losses of heterozygosity were significantly greater in high-grade adenomas than in low-grade adenomas, no significant differences in the lengths of CNAs were found between high-grade adenomas and IMAs. CONCLUSIONS Genomic alterations play an essential role in early colorectal carcinogenesis. CNAs in colorectal tumours provide new insights for evaluation of colorectal tumorigenesis.
Collapse
Affiliation(s)
- Makoto Eizuka
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, 19-1, Uchimaru, Morioka, 020-8505 Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, 19-1, Uchimaru, Morioka, 020-8505 Japan
| | - Wataru Habano
- Department of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, Morioka, Japan
| | - Noriyuki Uesugi
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, 19-1, Uchimaru, Morioka, 020-8505 Japan
| | - Yayoi Takahashi
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, 19-1, Uchimaru, Morioka, 020-8505 Japan
| | - Keisuke Kawasaki
- Division of Gastroenterology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Eiichiro Yamamoto
- Department of Molecular Biology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Takayuki Matsumoto
- Division of Gastroenterology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| |
Collapse
|