1
|
Ortega MA, Boaru DL, De Leon-Oliva D, De Castro-Martinez P, Minaya-Bravo AM, Casanova-Martín C, Barrena-Blázquez S, Garcia-Montero C, Fraile-Martinez O, Lopez-Gonzalez L, Saez MA, Alvarez-Mon M, Diaz-Pedrero R. The Impact of Klotho in Cancer: From Development and Progression to Therapeutic Potential. Genes (Basel) 2025; 16:128. [PMID: 40004457 PMCID: PMC11854833 DOI: 10.3390/genes16020128] [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: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Klotho, initially identified as an anti-aging gene, has been shown to play significant roles in cancer biology. Alongside α-Klotho, the β-Klotho and γ-Klotho isoforms have also been studied; these studies showed that Klotho functions as a potential tumor suppressor in many different cancers by inhibiting cancer cell proliferation, inducing apoptosis and modulating critical signaling pathways such as the Wnt/β-catenin and PI3K/Akt pathways. In cancers such as breast cancer, colorectal cancer, hepatocellular carcinoma, ovarian cancer, and renal cell carcinoma, reduced Klotho expression often correlates with a poor prognosis. In addition, Klotho's role in enhancing chemotherapy sensitivity and its epigenetic regulation further underscores its potential as a target for cancer treatments. This review details Klotho's multifaceted contributions to cancer suppression and its potential as a therapeutic target, enhancing the understanding of its significance in cancer treatment and prognoses.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Carlos Casanova-Martín
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, Central University Hospital of Defence—UAH Madrid, 28801 Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| |
Collapse
|
2
|
Chen Q, Zheng X, Cheng W, Li J. Landscape of targeted therapies for lung squamous cell carcinoma. Front Oncol 2024; 14:1467898. [PMID: 39544292 PMCID: PMC11560903 DOI: 10.3389/fonc.2024.1467898] [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: 07/24/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Lung cancer, a common type of malignant neoplasm, has seen significant advancements in the treatment of lung adenocarcinoma (LUAD). However, the management of lung squamous cell carcinoma (LSCC) continues to pose challenges. Traditional treatment methods for LSCC encompass surgical resection, chemotherapy, and radiotherapy. The introduction of targeted therapy and immunotherapy has greatly benefited LSCC patients, but issues such as limited immune response rates and adverse reactions persist. Therefore, gaining a deeper comprehension of the underlying mechanisms holds immense importance. This review provides an in-depth overview of classical signaling pathways and therapeutic targets, including the PI3K signaling pathway, CDK4/6 pathway, FGFR1 pathway and EGFR pathway. Additionally, we delve into alternative signaling pathways and potential targets that could offer new therapeutic avenues for LSCC. Lastly, we summarize the latest advancements in targeted therapy combined with immune checkpoint blockade (ICB) therapy for LSCC and discuss the prospects and challenges in this field.
Collapse
Affiliation(s)
- Qiuxuan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoshuo Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiting Cheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jian Li
- Institude of Experimental Immunology, University Clinic of Rheinische Friedrich-Wihelms-University, Bonn, Germany
| |
Collapse
|
3
|
Shen L, Wang C, Ren R, Liu X, Zhou D, Chen Y, Zhou Y, Lei J, Xiao Y, Zhang N, Zhao H, Li Y. Fibroblast growth factor receptor 4 deficiency in macrophages aggravates experimental colitis by promoting M1-polarization. Inflamm Res 2024; 73:1493-1510. [PMID: 38981913 DOI: 10.1007/s00011-024-01910-8] [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: 03/27/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVE AND DESIGN Compelling evidence indicates that dysregulated macrophages may play a key role in driving inflammation in inflammatory bowel disease (IBD). Fibroblast growth factor (FGF)-19, which is secreted by ileal enterocytes in response to bile acids, has been found to be significantly lower in IBD patients compared to healthy individuals, and is negatively correlated with the severity of diarrhea. This study aims to explore the potential impact of FGF19 signaling on macrophage polarization and its involvement in the pathogenesis of IBD. METHODS The dextran sulfate sodium (DSS)-induced mouse colitis model was utilized to replicate the pathology of human IBD. Mice were created with a conditional knockout of FGFR4 (a specific receptor of FGF19) in myeloid cells, as well as mice that overexpressing FGF19 specifically in the liver. The severity of colitis was measured using the disease activity index (DAI) and histopathological staining. Various techniques such as Western Blotting, quantitative PCR, flow cytometry, and ELISA were employed to assess polarization and the expression of inflammatory genes. RESULTS Myeloid-specific FGFR4 deficiency exacerbated colitis in the DSS mouse model. Deletion or inhibition of FGFR4 in bone marrow-derived macrophages (BMDMs) skewed macrophages towards M1 polarization. Analysis of transcriptome sequencing data revealed that FGFR4 deletion in macrophages significantly increased the activity of the complement pathway, leading to an enhanced inflammatory response triggered by LPS. Mechanistically, FGFR4-knockout in macrophages promoted complement activation and inflammatory response by upregulating the nuclear factor-κB (NF-κB)-pentraxin3 (PTX3) pathway. Additionally, FGF19 suppressed these pathways and reduced inflammatory response by activating FGFR4 in inflammatory macrophages. Liver-specific overexpression of FGF19 also mitigated inflammatory responses induced by DSS in vivo. CONCLUSION Our study highlights the significance of FGF19-FGFR4 signaling in macrophage polarization and the pathogenesis of IBD, offering a potential new therapeutic target for IBD.
Collapse
Affiliation(s)
- Luyao Shen
- The Second Affiliated Hospital & Yuying Children's Hospital, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Cong Wang
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Ran Ren
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Xudong Liu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Dongqin Zhou
- The Second Affiliated Hospital & Yuying Children's Hospital, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Yu Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yu Zhou
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Juan Lei
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yang Xiao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Nan Zhang
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China.
| | - Yongsheng Li
- The Second Affiliated Hospital & Yuying Children's Hospital, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
4
|
Wang X, Liu H, Wu G, Lu Y, Cui Y. Development and validation of a predictive model based on β-Klotho for head and neck squamous cell carcinoma. Sci Rep 2024; 14:17081. [PMID: 39048709 PMCID: PMC11269606 DOI: 10.1038/s41598-024-68130-2] [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: 01/16/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Head and neck epithelial tissue tumors may be identified as head and neck squamous cell carcinoma (HNSC). Numerous malignancies are encouraged by dysregulation of the FGF19-β-Klotho (KLB) axis in the tumor microenvironment. Using protein databases and RT-qPCR, we examined KLB expression in HNSC. In HNSC, higher KLB expression was linked to longer survival times and better prognoses. Furthermore, variations in drug susceptibility and immunological infiltration were noted according to KLB expression levels. These results underscore the importance of KLB in the course and management of HNSC by indicating that it may function as a possible prognostic marker and influence immunological and therapeutic responses in these individuals. Further study on HNSC is necessary to investigate KLB's potential as a therapeutic target and prognostic indicator.
Collapse
Affiliation(s)
- XiangXiu Wang
- Jinzhou Medical University, Jinzhou, 121000, China
- Department of Otorhinolaryngology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - HongWei Liu
- Cancer Hospital of China Medical University, Shenyang, 110042, China
| | - Gang Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yan Lu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Ying Cui
- Department of Otorhinolaryngology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
5
|
Mota J, Lima AMM, Gomes JIS, Souza de Andrade M, Brito HO, Silva MMAL, Faustino-Rocha AI, Oliveira PA, Lopes FF, Gil da Costa RM. Klotho in Cancer: Potential Diagnostic and Prognostic Applications. Diagnostics (Basel) 2023; 13:3357. [PMID: 37958253 PMCID: PMC10650206 DOI: 10.3390/diagnostics13213357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Klotho proteins, αKlotho, βKlotho, and γKlotho, exert tumor-suppressive activities via the fibroblast growth factor receptors and multiple cell-signaling pathways. There is a growing interest in Klotho proteins as potential diagnostic and prognostic biomarkers for multiple diseases. However, recent advances regarding their roles and potential applications in cancer remain disperse and require an integrated analysis. The present review analyzed research articles published between 2012 and 2022 in the Cochrane and Scopus scientific databases to study the role of Klotho in cancer and their potential as tools for diagnosing specific cancer types, predicting tumor aggressiveness and prognosis. Twenty-six articles were selected, dealing with acute myeloid leukemia and with bladder, breast, colorectal, esophageal, gastric, hepatocellular, ovarian, pancreatic, prostatic, pulmonary, renal, and thyroid cancers. αKlotho was consistently associated with improved prognosis and may be useful in estimating patient survival. A single study reported the use of soluble αKlotho levels in blood serum as a tool to aid the diagnosis of esophageal cancer. γKlotho was associated with increased aggressiveness of bladder, breast, and prostate cancer, and βKlotho showed mixed results. Further clinical development of Klotho-based assays will require careful identification of specific tumor subtypes where Klotho proteins may be most valuable as diagnostic or prognostic tools.
Collapse
Affiliation(s)
- Jucileide Mota
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Alice Marques Moreira Lima
- Health Sciences Center, State University of the Tocantins Region of Maranhão (UEMASUL), Imperatriz 6591-480, Brazil
| | - Jhessica I. S. Gomes
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Marcelo Souza de Andrade
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Haissa O. Brito
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
- Morphology Department, Federal University of Maranhão, São Luís 65085-580, Brazil
| | | | - Ana I. Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Fernanda F. Lopes
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Rui M. Gil da Costa
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- Associate Laboratory in Chemical Engineering, Faculty of Engineering (ALiCE), University of Porto, 4200-465 Porto, Portugal
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
- Health Research Network, Research Center of Portuguese Oncology Institute of Porto (CIIPOP/RISE@CIIPOP), 4200-072 Porto, Portugal
| |
Collapse
|
6
|
Wu T, Zhang Y, Han Q, Lu X, Cheng Y, Chen J, Sha J, Xia W. Klotho-beta attenuates Rab8a-mediated exosome regulation and promotes prostate cancer progression. Oncogene 2023; 42:2801-2815. [PMID: 37582861 DOI: 10.1038/s41388-023-02807-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023]
Abstract
Tumor-secreted exosomes have a wide range of effects on the growth, metastasis, and drug resistance of cancer cells. However, whether and how the molecular mechanisms that regulate the secretion of exosomes could affect tumor progression remains poorly understood. Klotho beta (KLB) has been reported dysregulated in prostate cancer, but its function remains unknown. Herein, we first determined that KLB was upregulated in prostate cancer and its expression level was positively correlated with prostate cancer malignant phenotype both in vitro and in vivo. Intriguingly, KLB overexpression could impair the release of exosomes and cause the intracellular accumulation of multivesicular bodies (MVBs) in prostate cancer cells. Mechanistically, KLB attenuated exosomes secretion through a Rab8a-dependent pathway. Rab8a was downregulated in KLB overexpressing cells whereas overexpression of Rab8a could rescue the impaired release of exosomes and attenuate the KLB-induced malignant phenotype of prostate cancer both in vitro and in vivo. Taken together, this study has unveiled the tumor-promoting role of KLB mediated by its regulation on exosomes secretion through a Rab8a-dependent mechanism. These findings could be exploited to develop novel theranostic targets for prostate cancer.
Collapse
Affiliation(s)
- Tingyu Wu
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanshuang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Han
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Lu
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yirui Cheng
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jiachen Chen
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Sha
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
7
|
Bedeschi M, Marino N, Cavassi E, Piccinini F, Tesei A. Cancer-Associated Fibroblast: Role in Prostate Cancer Progression to Metastatic Disease and Therapeutic Resistance. Cells 2023; 12:cells12050802. [PMID: 36899938 PMCID: PMC10000679 DOI: 10.3390/cells12050802] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers in European males. Although therapeutic approaches have changed in recent years, and several new drugs have been approved by the Food and Drug Administration (FDA), androgen deprivation therapy (ADT) remains the standard of care. Currently, PCa represents a clinical and economic burden due to the development of resistance to ADT, paving the way to cancer progression, metastasis, and to long-term side effects induced by ADT and radio-chemotherapeutic regimens. In light of this, a growing number of studies are focusing on the tumor microenvironment (TME) because of its role in supporting tumor growth. Cancer-associated fibroblasts (CAFs) have a central function in the TME because they communicate with prostate cancer cells, altering their metabolism and sensitivity to drugs; hence, targeted therapy against the TME, and, in particular, CAFs, could represent an alternative therapeutic approach to defeat therapy resistance in PCa. In this review, we focus on different CAF origins, subsets, and functions to highlight their potential in future therapeutic strategies for prostate cancer.
Collapse
Affiliation(s)
- Martina Bedeschi
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
- Correspondence: (M.B.); (A.T.); Tel.: +39-0543739932 (A.T.)
| | - Noemi Marino
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Elena Cavassi
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Filippo Piccinini
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Anna Tesei
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
- Correspondence: (M.B.); (A.T.); Tel.: +39-0543739932 (A.T.)
| |
Collapse
|
8
|
One-step metal affinity purification of recombinant hFGF19 without using tags. Protein Expr Purif 2023; 201:106186. [DOI: 10.1016/j.pep.2022.106186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
|
9
|
Justet A, Ghanem M, Boghanim T, Hachem M, Vasarmidi E, Jaillet M, Vadel A, Joannes A, Mordant P, Bonniaud P, Kolb M, Ling L, Cazes A, Mal H, Mailleux A, Crestani B. FGF19 is Downregulated in Idiopathic Pulmonary Fibrosis and Inhibits Lung Fibrosis in Mice. Am J Respir Cell Mol Biol 2022; 67:173-187. [PMID: 35549849 DOI: 10.1165/rcmb.2021-0246oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
IPF is a devastating lung disease with limited therapeutic possibilities. FGF19, an endocrine FGF, was recently shown to decrease liver fibrosis. To ask whether FGF19 had anti-fibrotic properties in the lung and decipher its effects on common features associated with lung fibrogenesis. We assessed, by Elisa, FGF19 levels in plasma and bronchoalveolar lavage fluids (BALF)obtained from controls and IPF patients. In vivo, using an intravenously administered adeno11 associated virus (AAV), we overexpressed FGF19 at the fibrotic phase of two experimental models of murine lung fibrosis and assessed its effect on lung morphology, lung collagen content, fibrosis markers and pro fibrotic mediator expression, at mRNA and protein levels. In vitro, we investigated whether FGF19 could modulate the TGFβ-induced differentiation of primary human lung fibroblast into myofibroblast and the apoptosis of murine alveolar type II cell. While FGF19 was not detected in BALF, FGF19 concentration was decreased in the plasma of IPF patients compared to controls. In vivo, the overexpression of FGF19 was associated with a marked decrease of lung fibrosis and fibrosis markers, with a decrease of pro fibrotic mediator expression and lung collagen content. In vitro, FGF19 decreased alveolar type 2 epithelial cell apoptosis through the decrease of the proapoptotic BIM protein expression and prevented TGF-ß induced myofibroblast differentiation through the inhibition of JNK phosphorylation. Altogether these data identify FGF19 as an anti-fibrotic molecule with a potential therapeutic interest in fibrotic lung disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Audrey Joannes
- INSERM U1085, IRSET Institut de Recherche sur la Santé, l'Environnement et le Travail, Université de Rennes-1, Rennes, France
| | - Pierre Mordant
- Assistance Publique - Hopitaux de Paris, 26930, Bichat Hospital, Department of Vascular and Thoracic Sugery, Paris, France.,INSERM, U1152, Paris, France.,Universite Paris Diderot UFR de Medecine Site Xavier-Bichat, 60152, Paris, France
| | - Philippe Bonniaud
- CHU Dijon-Bourgogne, Service de Pneumologie et Soins Intensifs Respiratoires, Dijon, France
| | - Martin Kolb
- McMaster University, Hamilton, Ontario, Canada
| | - Lei Ling
- NGM Biopharmaceuticals Inc, 200841, San Francisco, California, United States
| | | | | | - Arnaud Mailleux
- Inserm U700, Faculté de Médecine Paris 7, site X. Bichat, Paris, France
| | - Bruno Crestani
- AP-HP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Université Paris Diderot, Paris, France;
| |
Collapse
|
10
|
Agarwal S, Afaq F, Bajpai P, Kim H, Elkholy A, Behring M, Chandrashekar DS, Diffalha SA, Khushman M, Sugandha SP, Varambally S, Manne U. DCZ0415, a small-molecule inhibitor targeting TRIP13, inhibits EMT and metastasis via inactivation of the FGFR4/STAT3 axis and the Wnt/β-catenin pathway in colorectal cancer. Mol Oncol 2022; 16:1728-1745. [PMID: 35194944 PMCID: PMC9019876 DOI: 10.1002/1878-0261.13201] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 11/11/2022] Open
Abstract
Thyroid receptor-interacting protein 13 (TRIP13), a protein of the AAA-ATPase family, is upregulated in various human cancers, including colorectal cancer (CRC). This study focused on the inhibition of TRIP13-induced CRC progression and signalling by DCZ0415, a small molecule targeting TRIP13. It demonstrated potent antitumour activity in TRIP13-deregulated cancer cell lines, regardless of their p53, KRAS, BRAF, epidermal growth factor receptor or microsatellite instability status. The treatment of CRC cells with DCZ0415 resulted in decreased cell proliferation, induced cell cycle arrest in the G2-M phase and increased apoptosis. DCZ0415 diminished xenograft tumour growth and metastasis of CRC in immunocompromised mice. DCZ0415 reduced expression of fibroblast growth factor receptor 4 (FGFR4), signal transducer and activator of transcription 3 (STAT3), and proteins associated with the epithelial-mesenchymal transition and nuclear factor kappa B (NF-κB) pathways in cells and xenografts exhibiting high expression of TRIP13. Additionally, DCZ0415 decreased cyclin D1, β-catenin and T-cell factor 1, leading to the inactivation of the Wnt/β-catenin pathway. In a syngeneic CRC model, DCZ0415 treatment induced an immune response by decreasing PD1 and CTLA4 levels and increasing granzyme B, perforin and interferon gamma. In sum, DCZ04145 inhibits the TRIP13-FGFR4-STAT3 axis, inactivates NF-κB and Wnt/β-catenin signalling, activates antitumour immune response and reduces the progression and metastasis of CRC. This study provides a rationale to evaluate DCZ0415 clinically for the treatment of a subset of CRCs that exhibit dysregulated TRIP13 and FGFR4.
Collapse
Affiliation(s)
- Sumit Agarwal
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Farrukh Afaq
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Prachi Bajpai
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Hyung‐Gyoon Kim
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Amr Elkholy
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Michael Behring
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | | | - Sameer Al Diffalha
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Moh’d Khushman
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
- Department of MedicineDivision of Medical OncologyUniversity of Alabama at BirminghamALUSA
| | - Shajan P. Sugandha
- Department of MedicineDivision of GastroenterologyUniversity of Alabama at BirminghamALUSA
| | - Sooryanarayana Varambally
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Upender Manne
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| |
Collapse
|
11
|
Oshima K, Kato K, Ito Y, Daiko H, Nozaki I, Nakagawa S, Shibuya Y, Kojima T, Toh Y, Okada M, Hironaka S, Akiyama Y, Komatsu Y, Maejima K, Nakagawa H, Onuki R, Nagai M, Kato M, Kanato K, Kuchiba A, Nakamura K, Kitagawa Y. A prognostic biomarker study in patients with clinical stage I esophageal squamous cell carcinoma: JCOG0502-A1. Cancer Sci 2021; 113:1018-1027. [PMID: 34962019 PMCID: PMC8898710 DOI: 10.1111/cas.15251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/03/2022] Open
Abstract
We conducted genomic analyses of Japanese patients with stage I esophageal squamous cell carcinoma (ESCC) to investigate the frequency of genomic alterations and the association with survival outcomes. Biomarker analysis was conducted for patients with clinical stage T1bN0M0 ESCC enrolled in JCOG0502 (UMIN000000551). Whole-exome sequencing (WES) was performed using DNA extracted from formalin-fixed, paraffin-embedded tissue of ESCC and normal tissue or blood sample. Single nucleotide variants (SNVs), insertions/deletions (indels), and copy number alterations (CNAs) were identified. We then evaluated the associations between each gene alteration with a frequency ≥10% and progression-free survival (PFS) using a Cox regression model. We controlled for family-wise errors at 0.05 using the Bonferroni method. Among the 379 patients who were enrolled in JCOG0502, 127 patients were successfully analyzed using WES. The median patient age was 63 years (IQR, 57-67 years), and 78.0% of the patients ultimately underwent surgery. The 3-year PFS probability was 76.3%. We detected 20 genes with SNVs, indels, or amplifications with a frequency of ≥10%. Genomic alterations in FGF19 showed the strongest association with PFS with a borderline level of statistical significance of p = 0.00252 (Bonferroni-adjusted significance level is 0.0025). Genomic alterations in FGF4, MYEOV, CTTN, and ORAOV1 showed a marginal association with PFS (p < 0.05). These genomic alterations were all CNAs at chromosome 11q13.3. We have identified new genomic alterations associated with the poor efficacy of ESCC (T1bN0M0). These findings open avenues for the development of new potential treatments for patients with ESCC.
Collapse
Affiliation(s)
- Kotoe Oshima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo
| | - Yoshinori Ito
- Department of Radiation Oncology, Showa University School of Medicine, Tokyo
| | - Hiroyuki Daiko
- Esophageal Surgery Division, National Cancer Center Hospital, Tokyo
| | - Isao Nozaki
- Department of Gastroenterological Surgery, National Hospital Organization Shikoku Cancer Center, Matsuyama
| | - Satoru Nakagawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, Niigata
| | - Yuichi Shibuya
- Department of Gastroenterology Surgery, Kochi Health Sciences Center, Kochi
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa
| | - Yasushi Toh
- Department of Gastroenterological Surgery, National Hospital Organization Kyushu Cancer Center, Fukuoka
| | | | - Shuichi Hironaka
- Clinical Trial Promotion Department, Chiba Cancer Center, Chiba.,Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Yufu
| | - Yuji Akiyama
- Department of Surgery, Iwate Medical University, Iwate
| | - Yoshito Komatsu
- Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for integrative Medical Sciences, Yokohama
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for integrative Medical Sciences, Yokohama
| | - Ritsuko Onuki
- Division of Bioinformatics, Research Institute, National Cancer Center, Tokyo
| | - Momoko Nagai
- Division of Bioinformatics, Research Institute, National Cancer Center, Tokyo
| | - Mamoru Kato
- Division of Bioinformatics, Research Institute, National Cancer Center, Tokyo
| | - Keisuke Kanato
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo
| | - Aya Kuchiba
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo.,Graduate School of Health Innovation, Kanagawa University of Human Services, Kanagawa
| | - Kenichi Nakamura
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo
| |
Collapse
|
12
|
Chioni AM, Grose RP. Biological Significance and Targeting of the FGFR Axis in Cancer. Cancers (Basel) 2021; 13:5681. [PMID: 34830836 PMCID: PMC8616401 DOI: 10.3390/cancers13225681] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
The pleiotropic effects of fibroblast growth factors (FGFs), the widespread expression of all seven signalling FGF receptors (FGFRs) throughout the body, and the dramatic phenotypes shown by many FGF/R knockout mice, highlight the diversity, complexity and functional importance of FGFR signalling. The FGF/R axis is critical during normal tissue development, homeostasis and repair. Therefore, it is not surprising that substantial evidence also pinpoints the involvement of aberrant FGFR signalling in disease, including tumourigenesis. FGFR aberrations in cancer include mutations, gene fusions, and amplifications as well as corrupted autocrine/paracrine loops. Indeed, many clinical trials on cancer are focusing on targeting the FGF/FGFR axis, using selective FGFR inhibitors, nonselective FGFR tyrosine kinase inhibitors, ligand traps, and monoclonal antibodies and some have already been approved for the treatment of cancer patients. The heterogeneous tumour microenvironment and complexity of FGFR signalling may be some of the factors responsible for the resistance or poor response to therapy with FGFR axis-directed therapeutic agents. In the present review we will focus on the structure and function of FGF(R)s, their common irregularities in cancer and the therapeutic value of targeting their function in cancer.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Richard P. Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK;
| |
Collapse
|
13
|
Ferguson HR, Smith MP, Francavilla C. Fibroblast Growth Factor Receptors (FGFRs) and Noncanonical Partners in Cancer Signaling. Cells 2021; 10:1201. [PMID: 34068954 PMCID: PMC8156822 DOI: 10.3390/cells10051201] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence indicates that success of targeted therapies in the treatment of cancer is context-dependent and is influenced by a complex crosstalk between signaling pathways and between cell types in the tumor. The Fibroblast Growth Factor (FGF)/FGF receptor (FGFR) signaling axis highlights the importance of such context-dependent signaling in cancer. Aberrant FGFR signaling has been characterized in almost all cancer types, most commonly non-small cell lung cancer (NSCLC), breast cancer, glioblastoma, prostate cancer and gastrointestinal cancer. This occurs primarily through amplification and over-expression of FGFR1 and FGFR2 resulting in ligand-independent activation. Mutations and translocations of FGFR1-4 are also identified in cancer. Canonical FGF-FGFR signaling is tightly regulated by ligand-receptor combinations as well as direct interactions with the FGFR coreceptors heparan sulfate proteoglycans (HSPGs) and Klotho. Noncanonical FGFR signaling partners have been implicated in differential regulation of FGFR signaling. FGFR directly interacts with cell adhesion molecules (CAMs) and extracellular matrix (ECM) proteins, contributing to invasive and migratory properties of cancer cells, whereas interactions with other receptor tyrosine kinases (RTKs) regulate angiogenic, resistance to therapy, and metastatic potential of cancer cells. The diversity in FGFR signaling partners supports a role for FGFR signaling in cancer, independent of genetic aberration.
Collapse
Affiliation(s)
- Harriet R. Ferguson
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
| | - Michael P. Smith
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Science, Faculty of Biology Medicine and Health (FBMH), The University of Manchester, Manchester M13 9PT, UK;
- Manchester Breast Centre, Manchester Cancer Research Centre, The University of Manchester, Manchester M20 4GJ, UK
| |
Collapse
|
14
|
Giacomini A, Grillo E, Rezzola S, Ribatti D, Rusnati M, Ronca R, Presta M. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev 2020; 101:569-610. [PMID: 32730114 DOI: 10.1152/physrev.00005.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a family of proteins possessing paracrine, autocrine, or endocrine functions in a variety of biological processes, including embryonic development, angiogenesis, tissue homeostasis, wound repair, and cancer. Canonical FGFs bind and activate tyrosine kinase FGF receptors (FGFRs), triggering intracellular signaling cascades that mediate their biological activity. Experimental evidence indicates that FGFs play a complex role in the physiopathology of the prostate gland that ranges from essential functions during embryonic development to modulation of neoplastic transformation. The use of ligand- and receptor-deleted mouse models has highlighted the requirement for FGF signaling in the normal development of the prostate gland. In adult prostate, the maintenance of a functional FGF/FGFR signaling axis is critical for organ homeostasis and function, as its disruption leads to prostate hyperplasia and may contribute to cancer progression and metastatic dissemination. Dissection of the molecular landscape modulated by the FGF family will facilitate ongoing translational efforts directed toward prostate cancer therapy.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Domenico Ribatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| |
Collapse
|
15
|
Astolfi A, Pantaleo MA, Indio V, Urbini M, Nannini M. The Emerging Role of the FGF/FGFR Pathway in Gastrointestinal Stromal Tumor. Int J Mol Sci 2020; 21:E3313. [PMID: 32392832 PMCID: PMC7246647 DOI: 10.3390/ijms21093313] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal stromal tumors (GIST) are rare neoplasms of mesenchymal origin arising in the gastrointestinal tract. The vast majority are characterized by mutually exclusive activating mutations in KIT or Platelet-derived growth factor alpha (PDGFRA) receptors, or less frequently by succinate dehydrogenase complex (SDH) or NF1 inactivation, with very rare cases harboring mutant BRAF or RAS alleles. Approximately 5% of GISTs lack any of such mutations and are called quadruple wild-type (WT) GISTs. Recently, deregulated Fibroblast Growth Factor (FGF)/FGF-receptor (FGFR) signaling emerged as a relevant pathway driving oncogenic activity in different molecular subgroups of GISTs. This review summarizes all the current evidences supporting the key role of the FGF/FGFR pathway activation in GISTs, whereby either activating mutations, oncogenic gene fusions, or autocrine/paracrine signaling have been detected in quadruple WT, SDH-deficient, or KIT-mutant GISTs.
Collapse
Affiliation(s)
- Annalisa Astolfi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Maria Abbondanza Pantaleo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy
| | - Valentina Indio
- “Giorgio Prodi” Cancer Research Center, University of Bologna, 40138 Bologna, Italy;
| | - Milena Urbini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Margherita Nannini
- Medical Oncology Unit, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy;
| |
Collapse
|
16
|
Liu H, Zheng S, Hou X, Liu X, Du K, Lv X, Li Y, Yang F, Li W, Sui J. Novel Abs targeting the N-terminus of fibroblast growth factor 19 inhibit hepatocellular carcinoma growth without bile-acid-related side-effects. Cancer Sci 2020; 111:1750-1760. [PMID: 32061104 PMCID: PMC7226213 DOI: 10.1111/cas.14353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 02/05/2020] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common and particularly fatal form of cancer for which very few drugs are effective. The fibroblast growth factor 19 (FGF19) has been viewed as a driver of HCC development and a potential Ab target for developing novel HCC therapy. However, a previously developed anti‐FGF19 Ab disrupted FGF19’s normal regulatory function and caused severe bile‐acid‐related side‐effects despite of having potent antitumor effects in preclinical models. Here, we developed novel human Abs (G1A8 and HS29) that specifically target the N‐terminus of FGF19. Both Abs inhibited FGF19‐induced HCC cell proliferation in vitro and significantly suppressed HCC tumor growth in mouse models. Importantly, no bile‐acid‐related side effects were observed in preclinical cynomolgus monkeys. Fundamentally, our study demonstrates that it is possible to target FGF19 for anti‐HCC therapies without adversely affecting its normal bile acid regulatory function, and highlights the exciting promise of G1A8 or HS29 as potential therapy for HCC.
Collapse
Affiliation(s)
- Huisi Liu
- National Institute of Biological Sciences (NIBS), Beijing, China.,Peking University-Tsinghua University-National Institute of Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Peking University, Beijing, China
| | - Sanduo Zheng
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Xinfeng Hou
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Ximing Liu
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Kaixin Du
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Xueyuan Lv
- National Institute of Biological Sciences (NIBS), Beijing, China.,PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yulu Li
- National Institute of Biological Sciences (NIBS), Beijing, China.,Peking University-Tsinghua University-National Institute of Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Peking University, Beijing, China
| | - Fang Yang
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Wenhui Li
- National Institute of Biological Sciences (NIBS), Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Jianhua Sui
- National Institute of Biological Sciences (NIBS), Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
17
|
Lang L, Shull AY, Teng Y. Interrupting the FGF19-FGFR4 Axis to Therapeutically Disrupt Cancer Progression. Curr Cancer Drug Targets 2020; 19:17-25. [PMID: 29557750 DOI: 10.2174/1568009618666180319091731] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/01/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023]
Abstract
Coordination between the amplification of the fibroblast growth factor FGF19, overexpression of its corresponding receptor FGFR4, and hyperactivation of the downstream transmembrane enzyme β-klotho has been found to play pivotal roles in mediating tumor development and progression. Aberrant FGF19-FGFR4 signaling has been implicated in driving specific tumorigenic events including cancer cell proliferation, apoptosis resistance, and metastasis by activating a myriad of downstream signaling cascades. As an attractive target, several strategies implemented to disrupt the FGF19-FGFR4 axis have been developed in recent years, and FGF19-FGFR4 binding inhibitors are being intensely evaluated for their clinical use in treating FGF19-FGFR4 implicated cancers. Based on the established work, this review aims to detail how the FGF19-FGFR4 signaling pathway plays a vital role in cancer progression and why disrupting communication between FGF19 and FGFR4 serves as a promising therapeutic strategy for disrupting cancer progression.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology, Augusta University, Augusta, GA 30912, United States
| | - Austin Y Shull
- Department of Biology, Presbyterian College, Clinton, SC 29325, United States
| | - Yong Teng
- Department of Oral Biology, Augusta University, Augusta, GA 30912, United States.,Georgia Cancer Center, Augusta University, Augusta, GA 30912, United States.,Department of Biochemistry & Molecular Biology, Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
18
|
Li F, Li Z, Han Q, Cheng Y, Ji W, Yang Y, Lu S, Xia W. Enhanced autocrine FGF19/FGFR4 signaling drives the progression of lung squamous cell carcinoma, which responds to mTOR inhibitor AZD2104. Oncogene 2020; 39:3507-3521. [PMID: 32111983 PMCID: PMC7176586 DOI: 10.1038/s41388-020-1227-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/05/2020] [Accepted: 02/17/2020] [Indexed: 01/28/2023]
Abstract
Lung cancer occurrence and associated mortality ranks top in all countries. Despite the rapid development of targeted and immune therapies, many patients experience relapse within a few years. It is urgent to uncover the mechanisms that drive lung cancer progression and identify novel molecular targets. Our group has previously identified FGF19 as a prognostic marker and potential driver gene of lung squamous cell carcinomas (LSQ) in Chinese smoking patients. However, the underlying mechanism of how FGF19 promotes the progression of LSQ remains unclear. In this study, we characterized and confirmed that FGF19 serves as an oncogenic driver in LSQ development and progression, and reported that the amplification and high expression of FGF19 in LSQ was significantly associated with poor overall and progression-free survival. A higher serum level of FGF19 was found in lung cancer patients, which could also serve as a novel diagnostic index to screen lung cancer. Overproduction of FGF19 in LSQ cells markedly promoted cell growth, progression and metastasis, while downregulating FGF19 effectively inhibited LSQ progression in vitro and in vivo. Moreover, downregulating the receptor FGFR4 was also effective to suppress the growth and migration of LSQ cells. Since FGF19 could be induced by smoking or endoplasmic reticulum stress, to tackle the more malignant FGF19-overproducing LSQ, we reported for the first time that inhibiting mTOR pathway by using AZD2014 was effective and feasible. These findings have offered a new strategy by using anti-FGF19/FGFR4 therapy or mTOR-based therapy in FGF19-driven LSQ.
Collapse
Affiliation(s)
- Fan Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Han
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yirui Cheng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
19
|
Onishi K, Miyake M, Hori S, Onishi S, Iida K, Morizawa Y, Tatsumi Y, Nakai Y, Tanaka N, Fujimoto K. γ-Klotho is correlated with resistance to docetaxel in castration-resistant prostate cancer. Oncol Lett 2020; 19:2306-2316. [PMID: 32194730 PMCID: PMC7039116 DOI: 10.3892/ol.2020.11308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/16/2019] [Indexed: 01/09/2023] Open
Abstract
The Klotho (KL) gene was first identified as a potent aging suppressor. The KL family currently comprises of three proteins: α-Klotho (KLA), β-Klotho (KLB), and γ-Klotho (KLG). Many studies have shown that KLA and KLB participate in tumor progression or suppression, depending on the type of cancer; however, the relationship between KLG and prostate cancer has not yet been studied. Some studies have claimed that KL is correlated to sensitivity to chemotherapy. Here, we investigated the oncogenic potential of KLG in castration-resistant prostate cancer (CRPC). Immunohistochemical analysis using prostate biopsy specimens revealed that patients with high KLG expression in primary prostate cancer tissue had a significantly poor prognosis for overall survival. In addition, the prostate-specific antigen response rate after docetaxel (DTX) therapy in patients with high KLG expression was lower than that in patients with low KLG expression. To evaluate the potential of KLG as a therapeutic target in human prostate cancer, we generated a xenograft model of human CRPC cell line (PC-3) in male athymic mice. The animals were randomly divided into four groups as follows: i) control group (vehicle only); ii) DTX group (intraperitoneal administration); iii) small interfering RNA targeting KLG (KLG siRNA) group (intratumoral administration); and iv) a combination group (DTX plus KLG siRNA). After 3 weeks of treatment, the tumor weight and tumor Ki-67 labeling index were significantly lower in the KLG siRNA group and the combination group than in the control group. Sensitivity to DTX was increased upon treatment with KLG siRNA. These findings suggest that KLG expression in primary prostate cancer lesions is associated with resistance to DTX in CRPC and has potential as a diagnostic and therapeutic target for patients with CRPC.
Collapse
Affiliation(s)
- Kenta Onishi
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Makito Miyake
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Shunta Hori
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Sayuri Onishi
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kota Iida
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yoshihiro Tatsumi
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
20
|
Brominska B, Gabryel P, Jarmołowska-Jurczyszyn D, Janicka-Jedyńska M, Kluk A, Trojanowski M, Brajer-Luftmann B, Woliński K, Czepczyński R, Gut P, Bromiński G, Majewski P, Dyszkiewicz W, Ruchała M. Klotho expression and nodal involvement as predictive factors for large cell lung carcinoma. Arch Med Sci 2019; 15:1010-1016. [PMID: 31360195 PMCID: PMC6657266 DOI: 10.5114/aoms.2018.75889] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 05/22/2017] [Indexed: 01/30/2023] Open
Abstract
INTRODUCTION Klotho has been recently described as a carcinogenesis suppressor. Large cell neuroendocrine lung carcinoma (LCNEC) is a rare, highly malignant neoplasm. In the light of increasing incidence of neuroendocrine tumours, biomarkers predicting survival are needed. We consider that Klotho might be one. MATERIAL AND METHODS We analysed records of all patients diagnosed with LCNEC, atypical carcinoid and typical carcinoid operated on in our institution between 2007 and 2015. Initially, we found 134 cases. Forty-six specimens were unattainable and thus excluded from research. All patients diagnosed with LCNEC according to the WHO classification were included in the study. Immunohistochemical staining for Klotho was performed. We retrospectively reviewed patient charts and analysed multiple variables. RESULTS Positive staining for Klotho was present in 36 tissue specimens, while 12 patients were Klotho-negative. Survival length was significantly higher in Klotho-positive cases (p = 0.024), while advanced nodal status (N1 and N2) represented a marker of poor outcome (p = 0.011). In multivariate analysis, both Klotho presence (p = 0.015; HR = 0.37; 95% CI: 0.17-0.86) and nodal involvement (p = 0.007; HR = 3.04; 95% CI: 1.37-6.82) were independent prognostic factors. Tumour vessel invasion and visceral pleura infiltration were not associated with worse treatment results. Klotho presence predicted a favourable prognosis in these groups (p = 0.018; p = 0.007). CONCLUSIONS Our results suggest that Klotho might be a positive factor for predicting survival in LCNEC and nodal involvement a negative one. Thus, these two markers may assist in the selection of subjects with unfavourable prognosis and to personalise therapy regimens.
Collapse
Affiliation(s)
- Barbara Brominska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Piotr Gabryel
- Department of Thoracic Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Andrzej Kluk
- Department of Clinical Pathology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Trojanowski
- Department of Epidemiology and Cancer Prevention, Greater Poland Cancer Center, Poznan, Poland
| | - Beata Brajer-Luftmann
- Department of Pulmonology, Allergology and Respiratory Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kosma Woliński
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Rafał Czepczyński
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Gut
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Gabriel Bromiński
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Przemysław Majewski
- Department of Clinical Pathology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Dyszkiewicz
- Department of Thoracic Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
21
|
Abolghasemi M, Yousefi T, Maniati M, Qujeq D. The interplay of Klotho with signaling pathway and microRNAs in cancers. J Cell Biochem 2019; 120:14306-14317. [DOI: 10.1002/jcb.29022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Maryam Abolghasemi
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Department of Clinical Biochemistry, School of Medicine Babol University of Medical Sciences Babol Iran
- Student Research Committee Babol University of Medical Sciences Babol Iran
| | - Tooba Yousefi
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Department of Clinical Biochemistry, School of Medicine Babol University of Medical Sciences Babol Iran
- Student Research Committee Babol University of Medical Sciences Babol Iran
| | - Mahmood Maniati
- Assistant Professor of the English Department Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Department of Clinical Biochemistry, School of Medicine Babol University of Medical Sciences Babol Iran
| |
Collapse
|
22
|
Exploitation of phage display for the development of anti-cancer agents targeting fibroblast growth factor signaling pathways: New strategies to tackle an old challenge. Cytokine Growth Factor Rev 2019; 46:54-65. [DOI: 10.1016/j.cytogfr.2019.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/05/2019] [Accepted: 03/11/2019] [Indexed: 01/20/2023]
|
23
|
MicroRNA-520e restricts the proliferation and invasion of glioma cells through the downregulation of Wnt/β-catenin signaling by targeting fibroblast growth factor 19. Biochem Biophys Res Commun 2019; 511:619-625. [DOI: 10.1016/j.bbrc.2019.02.112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 12/22/2022]
|
24
|
Fibroblast Growth Factor Family in the Progression of Prostate Cancer. J Clin Med 2019; 8:jcm8020183. [PMID: 30720727 PMCID: PMC6406580 DOI: 10.3390/jcm8020183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 12/27/2022] Open
Abstract
Fibroblast growth factors (FGFs) and FGF receptors (FGFRs) play an important role in the maintenance of tissue homeostasis and the development and differentiation of prostate tissue through epithelial-stromal interactions. Aberrations of this signaling are linked to the development and progression of prostate cancer (PCa). The FGF family includes two subfamilies, paracrine FGFs and endocrine FGFs. Paracrine FGFs directly bind the extracellular domain of FGFRs and act as a growth factor through the activation of tyrosine kinase signaling. Endocrine FGFs have a low affinity of heparin/heparan sulfate and are easy to circulate in serum. Their biological function is exerted as both a growth factor binding FGFRs with co-receptors and as an endocrine molecule. Many studies have demonstrated the significance of these FGFs and FGFRs in the development and progression of PCa. Herein, we discuss the current knowledge regarding the role of FGFs and FGFRs—including paracrine FGFs, endocrine FGFs, and FGFRs—in the development and progression of PCa, focusing on the representative molecules in each subfamily.
Collapse
|
25
|
Li JR, Chiu KY, Ou YC, Wang SS, Chen CS, Yang CK, Ho HC, Cheng CL, Yang CR, Chen CC, Wang SC, Lin CY, Hung SC, Hsu CY, Chen CJ. Alteration in serum concentrations of FGF19, FGF21, and FGF23 in patients with urothelial carcinoma. Biofactors 2019; 45:62-68. [PMID: 30334297 DOI: 10.1002/biof.1460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/27/2018] [Accepted: 09/02/2018] [Indexed: 12/29/2022]
Abstract
Fibroblast growth factors (FGF) 19, 21, and 23 have been reported as functional factors in human metabolic diseases and malignancies. We performed a prospective survey to compare circulating FGF levels in urothelial carcinoma (UC) patients and normal controls. Between 2016 and 2017, 39 patients with UC of the urinary bladder or upper urinary tract who received surgical intervention were included. All the serum samples were obtained before surgeries. The control group included 28 healthy volunteers. Analysis of the circulating FGF19, 21, and 23 levels among all 67 subjects, as well as a subgroup analysis of the 39 UC patients were performed. The median levels of serum FGF19, 21, and 23 in the UC patients were 84.2, 505.3, and 117.6 pg/mL, respectively, which were statistically different from levels found in the healthy controls (P = 0.015, <0.001 and < 0.001, respectively). In the subgroup analysis, the FGF19 and FGF21 levels were significantly higher in end-stage renal disease UC patients, while FGF21 was also higher in the UC patients with cardiovascular diseases and history of recurrent UC. In the receiver operating characteristic (ROC) curve analysis, FGF19, 21, and 23 were all significant predictors of UC [area under the curve (AUC)] 0.674, P = 0.015; AUC 0.918, P < 0.001; AUC 0.897, P < 0.001, respectively). In UC patients, serum FGF19 level was significantly lower, while FGF21 and 23 were significantly higher, than respective levels in healthy controls. All three markers may serve as good predictors of UC occurrence, and FGF21 level was associated with disease recurrence. © 2018 BioFactors, 45(1):62-68, 2019.
Collapse
Affiliation(s)
- Jian-Ri Li
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medicine and Nursing, Hungkuang University, Taichung, Taiwan
| | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Yen-Chuan Ou
- Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shian-Shiang Wang
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Chuan-Su Chen
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Cheng-Kuang Yang
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hao-Chung Ho
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chen-Li Cheng
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chi-Rei Yang
- Department of Urology, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Che Chen
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shu-Chi Wang
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chia-Yen Lin
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Chun Hung
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chiann-Yi Hsu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| |
Collapse
|
26
|
Somm E, Jornayvaz FR. Fibroblast Growth Factor 15/19: From Basic Functions to Therapeutic Perspectives. Endocr Rev 2018; 39:960-989. [PMID: 30124818 DOI: 10.1210/er.2018-00134] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Discovered 20 years ago, fibroblast growth factor (FGF)19, and its mouse ortholog FGF15, were the first members of a new subfamily of FGFs able to act as hormones. During fetal life, FGF15/19 is involved in organogenesis, affecting the development of the ear, eye, heart, and brain. At adulthood, FGF15/19 is mainly produced by the ileum, acting on the liver to repress hepatic bile acid synthesis and promote postprandial nutrient partitioning. In rodents, pharmacologic doses of FGF19 induce the same antiobesity and antidiabetic actions as FGF21, with these metabolic effects being partly mediated by the brain. However, activation of hepatocyte proliferation by FGF19 has long been a challenge to its therapeutic use. Recently, genetic reengineering of the molecule has resolved this issue. Despite a global overlap in expression pattern and function, murine FGF15 and human FGF19 exhibit several differences in terms of regulation, molecular structure, signaling, and biological properties. As most of the knowledge originates from the use of FGF19 in murine models, differences between mice and humans in the biology of FGF15/19 have to be considered for a successful translation from bench to bedside. This review summarizes the basic knowledge concerning FGF15/19 in mice and humans, with a special focus on regulation of production, morphogenic properties, hepatocyte growth, bile acid homeostasis, as well as actions on glucose, lipid, and energy homeostasis. Moreover, implications and therapeutic perspectives concerning FGF19 in human diseases (including obesity, type 2 diabetes, hepatic steatosis, biliary disorders, and cancer) are also discussed.
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospitals, University of Geneva Medical School, Geneva, Switzerland
| | - François R Jornayvaz
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospitals, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
27
|
Secretome Screening Reveals Fibroblast Growth Factors as Novel Inhibitors of Viral Replication. J Virol 2018; 92:JVI.00260-18. [PMID: 29899088 DOI: 10.1128/jvi.00260-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/05/2018] [Indexed: 12/17/2022] Open
Abstract
Cellular antiviral programs can efficiently inhibit viral infection. These programs are often initiated through signaling cascades induced by secreted proteins, such as type I interferons, interleukin-6 (IL-6), or tumor necrosis factor alpha (TNF-α). In the present study, we generated an arrayed library of 756 human secreted proteins to perform a secretome screen focused on the discovery of novel modulators of viral entry and/or replication. The individual secreted proteins were tested for the capacity to inhibit infection by two replication-competent recombinant vesicular stomatitis viruses (VSVs) with distinct glycoproteins utilizing different entry pathways. Fibroblast growth factor 16 (FGF16) was identified and confirmed as the most prominent novel inhibitor of both VSVs and therefore of viral replication, not entry. Importantly, an antiviral interferon signature was completely absent in FGF16-treated cells. Nevertheless, the antiviral effect of FGF16 is broad, as it was evident on multiple cell types and also on infection by coxsackievirus. In addition, other members of the FGF family also inhibited viral infection. Thus, our unbiased secretome screen revealed a novel protein family capable of inducing a cellular antiviral state. This previously unappreciated role of the FGF family may have implications for the development of new antivirals and the efficacy of oncolytic virus therapy.IMPORTANCE Viruses infect human cells in order to replicate, while human cells aim to resist infection. Several cellular antiviral programs have therefore evolved to resist infection. Knowledge of these programs is essential for the design of antiviral therapeutics in the future. The induction of antiviral programs is often initiated by secreted proteins, such as interferons. We hypothesized that other secreted proteins may also promote resistance to viral infection. Thus, we tested 756 human secreted proteins for the capacity to inhibit two pseudotypes of vesicular stomatitis virus (VSV). In this secretome screen on viral infection, we identified fibroblast growth factor 16 (FGF16) as a novel antiviral against multiple VSV pseudotypes as well as coxsackievirus. Subsequent testing of other FGF family members revealed that FGF signaling generally inhibits viral infection. This finding may lead to the development of new antivirals and may also be applicable for enhancing oncolytic virus therapy.
Collapse
|
28
|
Abstract
The prostate is a male exocrine gland that secretes components of the seminal fluid. In men, prostate tumors are one of the most prevalent cancers. Studies on the development of the prostate have given a better understanding of the processes and genes that are important in the formation of this organ and have provided insights into the mechanisms of prostate tumorigenesis. These developmental studies have provided evidence that some of the genes and signaling pathways involved in development are reactivated or deregulated during prostate cancer. The prostate goes through a number of different stages during organogenesis, which include organ specification, epithelial budding, branching morphogenesis, canalization, and cytodifferentiation. During development, these processes are tightly regulated, many of which are controlled by the male hormone androgens. The majority of prostate tumors remain hormone regulated, and antiandrogen therapy is a first-line therapy, highlighting the important link between prostate organogenesis and cancer. In this review, we describe some of the data on genes that have important roles during prostate development that also have strong evidence linking them to prostate cancer.
Collapse
Affiliation(s)
- Jeffrey C Francis
- Division of Cancer Biology, Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - Amanda Swain
- Division of Cancer Biology, Institute of Cancer Research, London SW3 6JB, United Kingdom
| |
Collapse
|
29
|
Liu Z, Zhang H, Ding S, Qi S, Liu S, Sun D, Dong W, Yin L, Li M, Zhao X, Lu J. βKlotho inhibits androgen/androgen receptor‑associated epithelial‑mesenchymal transition in prostate cancer through inactivation of ERK1/2 signaling. Oncol Rep 2018; 40:217-225. [PMID: 29749458 PMCID: PMC6059743 DOI: 10.3892/or.2018.6399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is reported to have intimate crosstalk with androgen receptor (AR) signaling in prostate cancer (PCa) and is known to be responsible for castration resistance. Fibroblast growth factor/receptor (FGF/FGFR) signaling is also involved in tumor progression and EMT in multiple tissues. Several studies have investigated the role of βKlotho, an FGF/FGFR signaling co-receptor in tumorigenesis. However, its role in PCa remains unknown. In the present study, the role of androgen in the EMT of PCa cells was examined by western blotting. The expression of βKlotho was examined in prostate cells and PCa tissues by western blotting and immunohistochemistry, respectively. The biological role of βKlotho was revealed by a series of functional in vitro and in vivo studies. We determined that βKlotho expression was significantly decreased in PCa tissues compared with benign prostatic hyperplasia (BPH) tissues, and low βKlotho expression was associated with a high Gleason score of PCa. βKlotho overexpression inhibited the viability, migration, and androgen/AR-associated EMT of PCa cells through the inactivation of ERK1/2 signaling. Notably, βKlotho overexpression inhibited prostate tumor growth and EMT in vivo. Knockdown of βKlotho produced the opposite effects. In conclusion, βKlotho inhibits EMT and plays a tumor-suppressive role in PCa, linking FGF/FGFR/βKlotho signaling to the regulation of PCa progression.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Sentai Ding
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shasha Qi
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shuai Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Dingqi Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Wei Dong
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Lei Yin
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Mingjiang Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xingbo Zhao
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jiaju Lu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
30
|
Gamma-Klotho exhibits multiple roles in tumor growth of human bladder cancer. Oncotarget 2018; 9:19508-19524. [PMID: 29731962 PMCID: PMC5929405 DOI: 10.18632/oncotarget.24628] [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] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/24/2018] [Indexed: 01/08/2023] Open
Abstract
Alpha-Klotho (KLα) and beta-Klotho (KLβ) have recently been reported to correlate with cancer prognosis in some malignancies and we previously reported the association between KLα, KLβ, and urothelial carcinoma of the bladder (UCB), indicating that KLβ acts as a tumor promoter. However, the association between gamma-Klotho (KLγ) and cancer prognosis remains unclear. In the present study, we evaluated the association between KLγ and UCB. To evaluate the effect of KLγ on human bladder cancer cell lines in vitro assays were performed. Exogenous KLγ increased the ability of human bladder cancer cells to proliferate, migrate, invade, form colonies, and provide anchorage-independent growth potential. In in vivo assays, eighteen mice bearing xenografts inoculated using UM-UC-3, were randomly divided into three groups and treated with a small interfering RNA (siRNA) by intratumoral administration once a week for four weeks. Knockdown of KLγ with siRNA led to a dramatic change in tumor growth and suggested that KLγ had effects on tumor growth, including promotion of cell proliferation, inhibition of apoptosis, and enhancement of the epithelial-mesenchymal transition. To confirm the study, human tissue samples were used and patients were divided into two groups according to KLγ expression level. High expression of KLγ was significantly associated with higher stage and grade cancer and the presence of lymphovascular invasion compared to patients with lower expression of KLγ. Our results suggest that KLγ plays an important role in tumor invasion and progression and these results may lead to the development of new therapies and diagnostic methods for UCB.
Collapse
|
31
|
Feng S, Shao L, Castro P, Coleman I, Nelson PS, Smith PD, Davies BR, Ittmann M. Combination treatment of prostate cancer with FGF receptor and AKT kinase inhibitors. Oncotarget 2018; 8:6179-6192. [PMID: 28008155 PMCID: PMC5351622 DOI: 10.18632/oncotarget.14049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/13/2016] [Indexed: 12/25/2022] Open
Abstract
Activation of the PI3K/AKT pathway occurs in the vast majority of advanced prostate cancers (PCas). Activation of fibroblast growth factor receptor (FGFR) signaling occurs in a wide variety of malignancies, including PCa. RNA-Seq of castration resistant PCa revealed expression of multiple FGFR signaling components compatible with FGFR signaling in all cases, with multiple FGF ligands expressed in 90% of cases. Immunohistochemistry confirmed FGFR signaling in the majority of xenografts and advanced PCas. AZD5363, an AKT kinase inhibitor and AZD4547, a FGFR kinase inhibitor are under active clinical development. We therefore sought to determine if these two drugs have additive effects in PCa models. The effect of both agents, singly and in combination was evaluated in a variety of PCa cell lines in vitro and in vivo. All cell lines tested responded to both drugs with decreased invasion, soft agar colony formation and growth in vivo, with additive effects seen with combination treatment. Activation of the FGFR, AKT, ERK and STAT3 pathways was examined in treated cells. AZD5363 inhibited AKT signaling and increased FGFR1 signaling, which partially compensated for decreased AKT kinase activity. While AZD4547 could effectively block the ERK pathway, combination treatment was needed to completely block STAT3 activation. Thus combination treatment with AKT and FGFR kinase inhibitors have additive effects on malignant phenotypes in vitro and in vivo by inhibiting multiple signaling pathways and mitigating the compensatory upregulation of FGFR signaling induced by AKT kinase inhibition. Our studies suggest that co-targeting these pathways may be efficacious in advanced PCa.
Collapse
Affiliation(s)
- Shu Feng
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Department of Veterans Affairs Medical Center Baylor College of Medicine, Houston, 77030, TX, USA
| | - Longjiang Shao
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Department of Veterans Affairs Medical Center Baylor College of Medicine, Houston, 77030, TX, USA
| | - Patricia Castro
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Department of Veterans Affairs Medical Center Baylor College of Medicine, Houston, 77030, TX, USA
| | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Paul D Smith
- Oncology iMED, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Barry R Davies
- Oncology iMED, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Michael Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Department of Veterans Affairs Medical Center Baylor College of Medicine, Houston, 77030, TX, USA
| |
Collapse
|
32
|
Shao L, Wang J, Karatas OF, Feng S, Zhang Y, Creighton CJ, Ittmann M. Fibroblast growth factor receptor signaling plays a key role in transformation induced by the TMPRSS2/ERG fusion gene and decreased PTEN. Oncotarget 2018; 9:14456-14471. [PMID: 29581856 PMCID: PMC5865682 DOI: 10.18632/oncotarget.24470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/03/2018] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the most common visceral malignancy and the second leading cause of cancer deaths in US men. Correlative studies in human prostate cancers reveal a frequent association of the TMPRSS2/ERG (TE) fusion gene with loss of PTEN and studies in mouse models reveal that ERG expression and PTEN loss synergistically promote prostate cancer progression. To determine the mechanism by which ERG overexpression and PTEN loss leads to transformation, we overexpressed the TE fusion gene and knocked down PTEN in an immortalized but non-transformed prostate epithelial cell line. We show that ERG overexpression in combination with PTEN loss can transform these immortalized but non-tumorigenic cells, while either alteration alone was not sufficient to fully transform these cells. Expression microarray analysis revealed extensive changes in gene expression in cells expressing the TE fusion with loss of PTEN. Among these gene expression changes was increased expression of multiple FGF ligands and receptors. We show that activation of fibroblast growth factor receptor signaling plays a key role in transformation induced by TE fusion gene expression in association with PTEN loss. In addition, in vitro and in silico analysis reveals PTEN loss is associated with widespread increases in FGF ligands and receptors in prostate cancer. Inhibitors of FGF receptor signaling are currently entering the clinic and our results suggests that FGF receptor signaling is a therapeutic target in cancers with TE fusion gene expression and PTEN loss.
Collapse
Affiliation(s)
- Longjiang Shao
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Jianghua Wang
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Omer Faruk Karatas
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Shu Feng
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Yiqun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michael Ittmann
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| |
Collapse
|
33
|
Motylewska E, Stępień T, Borkowska M, Kuzdak K, Siejka A, Komorowski J, Stępień H, Ławnicka H. Alteration in the serum concentrations of FGF19, FGFR4 and βKlotho in patients with thyroid cancer. Cytokine 2018; 105:32-36. [PMID: 29438906 DOI: 10.1016/j.cyto.2018.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION βKlotho (βKL) is known to act as co-receptor for fibroblast growth factor receptor 4 (FGFR4) which is the main cognate receptor for fibroblast growth factor 19 (FGF19). Dysregulation of this FGF19/FGFR4/βKL signaling axis has been implicated in the pathogenesis of several cancers. However, its role in the pathogenesis of thyroid cancer has not been determined. MATERIALS AND METHODS The aim of this study was to assess FGF19, FGFR4 and βKL concentrations in a group of 36 patients with papillary thyroid cancer (PTC), 11 patients with follicular thyroid cancer (FTC), 9 patients with anaplastic thyroid cancer (ATC) and a group of 19 subjects with multinodular nontoxic goiter (MNG). The control group consisted of 20 healthy volunteers. Serum FGF19, FGFR4 and βKL concentrations were measured using specific ELISA methods. RESULTS Significantly lower concentrations of βKL and higher concentrations of FGF19 were found in patients with PTC, FTC and ATC as compared with MNG group and controls. An elevation of FGFR4 serum concentration was observed in all thyroid cancer groups in comparison to MNG group and controls; however, in FTC group it was statistically insignificant. A positive correlation was found between βKL and FGFR4 concentrations in PTC patients. The levels of βKL, FGF19 and FGFR4 did not differ significantly between MNG group and healthy controls. CONCLUSIONS Our results indicate that a disrupted FGF19/FGFR4/βKL signaling pathway may play a role in the development of thyroid cancers. However, further studies are needed to elucidate the molecular mechanism of the neoplastic transition of thyroid epithelial cells.
Collapse
Affiliation(s)
- Ewelina Motylewska
- Department of Immunoendocrinology, Chair of Endocrinology, Medical University of Lodz, Sterlinga 3, 91-425 Lodz, Poland.
| | - Tomasz Stępień
- Clinic of Endocrinological and General Surgery, Chair of Endocrinology, Medical University of Lodz, Pabianicka 62, 93-513 Lodz, Poland
| | - Magdalena Borkowska
- Clinic of Endocrinological and General Surgery, Chair of Endocrinology, Medical University of Lodz, Pabianicka 62, 93-513 Lodz, Poland
| | - Krzysztof Kuzdak
- Clinic of Endocrinological and General Surgery, Chair of Endocrinology, Medical University of Lodz, Pabianicka 62, 93-513 Lodz, Poland
| | - Agnieszka Siejka
- Clinic of Endocrinology, Chair of Endocrinology, Medical University of Lodz, Sterlinga 3, 91-425 Lodz, Poland
| | - Jan Komorowski
- Clinic of Endocrinology, Chair of Endocrinology, Medical University of Lodz, Sterlinga 3, 91-425 Lodz, Poland
| | - Henryk Stępień
- Department of Immunoendocrinology, Chair of Endocrinology, Medical University of Lodz, Sterlinga 3, 91-425 Lodz, Poland
| | - Hanna Ławnicka
- Department of Immunoendocrinology, Chair of Endocrinology, Medical University of Lodz, Sterlinga 3, 91-425 Lodz, Poland
| |
Collapse
|
34
|
Teng Y, Zhao H, Gao L, Zhang W, Shull AY, Shay C. FGF19 Protects Hepatocellular Carcinoma Cells against Endoplasmic Reticulum Stress via Activation of FGFR4-GSK3β-Nrf2 Signaling. Cancer Res 2017; 77:6215-6225. [PMID: 28951455 DOI: 10.1158/0008-5472.can-17-2039] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/07/2017] [Accepted: 09/15/2017] [Indexed: 01/03/2023]
Abstract
The tumor microenvironment induces endoplasmic reticulum (ER) stress in tumor cells, an event that can promote progression, but it is unknown how tumor cells adapt to this stress. In this study, we show that the fibroblast growth factor FGF19, a gene frequently amplified in hepatocellular carcinoma (HCC), facilitates a survival response to ER stress. Levels of FGF19 expression were increased in stressed HCC cells in culture and in a mouse xenograft model. Induction of ER stress required the transcription factor ATF4, which directly bound the FGF19 promoter. In cells where ER stress was induced, FGF19 overexpression promoted HCC cell survival and increased resistance to apoptosis, whereas FGF19 silencing counteracted these effects. Mechanistic investigations implicated glycogen synthase kinase-3β (GSK3β) in regulating nuclear accumulation of the stress-regulated transcription factor Nrf2 activated by FGF19. Our findings show how FGF19 provides a cytoprotective role against ER stress by activating a FGFR4-GSK3β-Nrf2 signaling cascade, with implications for targeting this signaling node as a candidate therapeutic regimen for HCC management. Cancer Res; 77(22); 6215-25. ©2017 AACR.
Collapse
Affiliation(s)
- Yong Teng
- Department of Oral Biology, Augusta University, Augusta, Georgia. .,Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Huakan Zhao
- School of Life Sciences, Chongqing University, Chongqing, China.,Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lixia Gao
- Department of Oral Biology, Augusta University, Augusta, Georgia
| | - Wenfa Zhang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Austin Y Shull
- Department of Biology, Presbyterian College, Clinton, South Carolina
| | - Chloe Shay
- Emory Children's Center, Emory University, Atlanta, Georgia
| |
Collapse
|
35
|
Olauson H, Mencke R, Hillebrands JL, Larsson TE. Tissue expression and source of circulating αKlotho. Bone 2017; 100:19-35. [PMID: 28323144 DOI: 10.1016/j.bone.2017.03.043] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
αKlotho (Klotho), a type I transmembrane protein and a coreceptor for Fibroblast Growth Factor-23, was initially thought to be expressed only in a limited number of tissues, most importantly the kidney, parathyroid gland and choroid plexus. Emerging data may suggest a more ubiquitous Klotho expression pattern which has prompted reevaluation of the restricted Klotho paradigm. Herein we systematically review the evidence for Klotho expression in various tissues and cell types in humans and other mammals, and discuss potential reasons behind existing conflicting data. Based on current literature and tissue expression atlases, we propose a classification of tissues into high, intermediate and low/absent Klotho expression. The functional relevance of Klotho in organs with low expression levels remain uncertain and there is currently limited data on a role for membrane-bound Klotho outside the kidney. Finally, we review the evidence for the tissue source of soluble Klotho, and conclude that the kidney is likely to be the principal source of circulating Klotho in physiology.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Rik Mencke
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Clayton NS, Wilson AS, Laurent EP, Grose RP, Carter EP. Fibroblast growth factor-mediated crosstalk in cancer etiology and treatment. Dev Dyn 2017; 246:493-501. [PMID: 28470714 DOI: 10.1002/dvdy.24514] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/21/2017] [Accepted: 04/25/2017] [Indexed: 12/26/2022] Open
Abstract
It is becoming increasingly evident that multiple cell types within the tumor work together to drive tumour progression and impact on both the response to therapy and the dissemination of tumour cells throughout the body. Fibroblast growth factor signalling (FGF) is perturbed in a number of tumors, serving to drive tumor cell proliferation and migration, but also has a central role in orchestrating the plethora of cells that comprise the tumor microenvironment. This review focuses on how this family of signalling molecules can influence the interactions between tumor cells and their surrounding environment. Unraveling the complexities of FGF signalling between the distinct cell types of a tumor may identify additional opportunities for FGF-targeted compounds in therapy and could help combat drug resistance. Developmental Dynamics 246:493-501, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- N S Clayton
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - A S Wilson
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - E P Laurent
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - R P Grose
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - E P Carter
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
37
|
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate numerous cellular processes. Deregulation of FGFR signalling is observed in a subset of many cancers, making activated FGFRs a highly promising potential therapeutic target supported by multiple preclinical studies. However, early-phase clinical trials have produced mixed results with FGFR-targeted cancer therapies, revealing substantial complexity to targeting aberrant FGFR signalling. In this Review, we discuss the increasing understanding of the differences between diverse mechanisms of oncogenic activation of FGFR, and the factors that determine response and resistance to FGFR targeting.
Collapse
Affiliation(s)
- Irina S Babina
- Breast Cancer Now Research Centre, Institute of Cancer Research, London SW3 6JB, UK
| | - Nicholas C Turner
- Breast Cancer Now Research Centre, Institute of Cancer Research, London SW3 6JB, UK
- Breast Unit, The Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
| |
Collapse
|
38
|
Zhou X, Fang X, Jiang Y, Geng L, Li X, Li Y, Lu K, Li P, Lv X, Wang X. Klotho, an anti-aging gene, acts as a tumor suppressor and inhibitor of IGF-1R signaling in diffuse large B cell lymphoma. J Hematol Oncol 2017; 10:37. [PMID: 28153033 PMCID: PMC5288890 DOI: 10.1186/s13045-017-0391-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/03/2017] [Indexed: 01/13/2023] Open
Abstract
Background Klotho, is a transmembrane protein, performs as a circulating hormone and upstream modulator of the insulin-like growth factor-1 receptor (IGF-1R), fibroblast growth factor (FGF), and Wnt signaling pathways. These pathways are involved in the development and progression of B cell lymphoma. We aimed to explore the expression pattern and functional mechanism of Klotho in diffuse large B cell lymphoma (DLBCL). Methods Immunohistochemistry (IHC) and western blotting were performed to detect the expression level of Klotho in DLBCL patients and cell lines. Tumor suppressive effect of Klotho was determined by both in vitro and in vivo studies. Signaling pathway activity was assessed by western blotting. Results Remarkable lower expression levels of Klotho were observed in DLBCL patients and cell lines. Enforced expression of Klotho could significantly induce cell apoptosis and inhibit tumor growth in DLBCL. Upregulation of Klotho resulted in declined activation of IGF-1R signaling, accompanied with decreased phosphorylation of its downstream targets, including AKT and ERK1/2. Moreover, xenograft model treated with either Klotho overexpression vector or recombinant human Klotho administration presented restrained tumor growth and lower Ki67 staining. Conclusions Our findings establish that Klotho performs as a tumor suppressor and modulator of IGF-1R signaling in DLBCL. Targeting Klotho may provide novel strategies for future therapeutic intervention.
Collapse
Affiliation(s)
- Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Lingyun Geng
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Xinyu Li
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Ying Li
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Peipei Li
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Xiao Lv
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China. .,Shandong University School of Medicine, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
39
|
Wang S, Zhao D, Tian R, Shi H, Chen X, Liu W, Wei L. FGF19 Contributes to Tumor Progression in Gastric Cancer by Promoting Migration and Invasion. Oncol Res 2016; 23:197-203. [PMID: 27053348 PMCID: PMC7838742 DOI: 10.3727/096504016x14537290676919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Gastric cancer is the fourth most common type of cancer and second leading cause of cancer-related death in the world. Since patients are often diagnosed at a late stage, very few effective therapies are left in the arsenal. FGF19, as a hormone, has been reported to promote tumor growth in various types of cancer; however, its function in gastric cancer remains unknown. In the current study, we showed that FGF19 is overexpressed in gastric cancer and is associated with depth of invasion, lymph node metastasis, and TNM stage. In addition, in vitro experiments demonstrated that FGF19 is able to enhance migration and invasion abilities of gastric cancer cells. Given its great potency in gastric cancer progression, FGF19 may be an effective target of treatment for advanced gastric cancer patients.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Clinical Oncology, The Central Hospital of Taian, Taian, Shandong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Hori S, Miyake M, Onishi S, Tatsumi Y, Morizawa Y, Nakai Y, Anai S, Tanaka N, Fujimoto K. Clinical significance of α‑ and β‑Klotho in urothelial carcinoma of the bladder. Oncol Rep 2016; 36:2117-25. [PMID: 27573985 DOI: 10.3892/or.2016.5053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/05/2016] [Indexed: 11/06/2022] Open
Abstract
Non-muscle invasive bladder cancer (NMIBC) accounts for ~70% of all bladder cancers. One of the serious clinical issues related to the management of NMIBC is that it has significant potential to progress to muscle invasive bladder cancer (MIBC) after initial treatments. α‑Klotho (KLα), originally identified as an anti‑aging gene, has recently been reported to have antitumor effects in various malignancies. In contrast, β‑Klotho (KLβ) has been reported to have protumoral functions. However, the associations between KLα/KLβ and the biological behavior of urothelial carcinoma remain unclear. In the present study, we evaluated the association between clinicopathological background factors of NMIBC and the expression levels of KLα or KLβ. A high expression level of KLβ, but not KLα, was an independent predictive factor of short progression‑free survival for NMIBC. An elevated level of KLβ correlated with a higher incidence of lymphovascular invasion (LVI). We added in vitro assays using human bladder cancer cell lines to investigate the role of KLβ. Treatment with exogenous KLβ protein increased the proliferation, migration, transendothelial migration abilities and anchorage‑independent growth of the cell lines. In addition, the KLβ concentration in voided urine samples obtained before initial transurethral surgery was quantitated with enzyme‑linked immunosorbent assay (ELISA). The urine KLβ concentration was found to be higher in patients with bladder cancer than that in healthy volunteers. Our results suggest that KLβ plays important roles in tumor invasion and progression, and its concentration may be a valuable urine‑based marker for the detection of bladder cancer.
Collapse
Affiliation(s)
- Shunta Hori
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Makito Miyake
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Sayuri Onishi
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Yoshihiro Tatsumi
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Satoshi Anai
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, Kashihara, Nara 634‑8522, Japan
| |
Collapse
|
41
|
Abstract
Prostate cancer is the most common cancer in US men and the second leading cause of cancer deaths. Fibroblast growth factor 23 (FGF23) is an endocrine FGF, normally expressed by osteocytes, which plays a critical role in phosphate homeostasis via a feedback loop involving the kidney and vitamin D. We now show that FGF23 is expressed as an autocrine growth factor in all prostate cancer cell lines tested and is present at increased levels in prostate cancer tissues. Exogenous FGF23 enhances proliferation, invasion and anchorage independent growth in vitro while FGF23 knockdown in prostate cancer cell lines decreases these phenotypes. FGF23 knockdown also decreases tumor growth in vivo. Given that classical FGFs and FGF19 are also increased in prostate cancer, we analyzed expression microarrays hybridized with RNAs from of LNCaP cells stimulated with FGF2, FGF19 or FGF23. The different FGF ligands induce overlapping as well as unique patterns of gene expression changes and thus are not redundant. We identified multiple genes whose expression is altered by FGF23 that are associated with prostate cancer initiation and progression. Thus FGF23 can potentially also act as an autocrine, paracrine and/or endocrine growth factor in prostate cancer that can promote prostate cancer progression.
Collapse
|
42
|
Zhao H, Lv F, Liang G, Huang X, Wu G, Zhang W, Yu L, Shi L, Teng Y. FGF19 promotes epithelial-mesenchymal transition in hepatocellular carcinoma cells by modulating the GSK3β/β- catenin signaling cascade via FGFR4 activation. Oncotarget 2016; 7:13575-13586. [PMID: 26498355 PMCID: PMC4924662 DOI: 10.18632/oncotarget.6185] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/04/2015] [Indexed: 12/12/2022] Open
Abstract
Compelling evidence suggests that the epithelial-mesenchymal transition (EMT) correlates with aggressiveness of tumors and poor survival. FGF19 has been shown to be involved in EMT in cholangiocarcinoma and colorectal cancer, however, molecular mechanisms underlying FGF19-induced EMT process in hepatocellular carcinoma (HCC) remain largely unknown. Here, we show the expression of FGF19 is significantly elevated and negatively associated with the expression of E-cadherin in HCC tissues and cell lines. Ectopic FGF19 expression promotes EMT and invasion in epithelial-like HCC cells through repression of E-cadherin expression, whereas FGF19 knockdown enhances E-cadherin expression and hence diminishes EMT traits in mesenchymal-like HCC cells, suggesting FGF19 exerts its tumor progressing functions as an EMT inducer. Interestingly, depletion of FGF19 cannot abrogate EMT traits in the presence of GSK3β inhibitors. Furthermore, FGF19-induced EMT can be markedly attenuated when FGFR4 is knocked out. These observations clearly indicate that FGFR4/GSK3β/β-catenin axis may play a pivotal role in FGF19-induced EMT in HCC cells. As FGF19 and its specific receptor FGFR4 are frequently amplified in HCC cells, selective targeting this signaling node may lend insights into a potential effective therapeutic approach for blocking metastasis of HCC.
Collapse
MESH Headings
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Case-Control Studies
- Cell Movement
- Cell Proliferation
- Epithelial-Mesenchymal Transition
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Glycogen Synthase Kinase 3 beta/genetics
- Glycogen Synthase Kinase 3 beta/metabolism
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Prognosis
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Huakan Zhao
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Fenglin Lv
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guizhao Liang
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Xiaobin Huang
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Gang Wu
- Third Affiliated Hospital, Third Military Medical University, Chongqing, PR China
| | - Wenfa Zhang
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Le Yu
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Lei Shi
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Teng
- School of Life Sciences and School of Bioengineering, Chongqing University, Chongqing, PR China
| |
Collapse
|
43
|
Shi S, Zhang Q, Xia Y, You B, Shan Y, Bao L, Li L, You Y, Gu Z. Mesenchymal stem cell-derived exosomes facilitate nasopharyngeal carcinoma progression. Am J Cancer Res 2016; 6:459-472. [PMID: 27186416 PMCID: PMC4859673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/08/2016] [Indexed: 06/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs), which are capable of differentiating into multiple cell types, are reported to exert multiple effects on tumor development. However, the relationship between MSCs and nasopharyngeal carcinoma (NPC) cells remains unclear. Exosomes are small membrane vesicles that can be released by several cell types, including MSCs. Exosomes, which can carry membrane and cytoplasmic constituents, have been described as participants in a novel mechanism of cell-to-cell communication. In the present study, we investigated the mechanisms underlying the interaction between MSCs and NPC cells. The data showed that MSCs secreted 40-100 nm heterogeneous small vesicles, which were defined as exosomes. Incubation of NPC cells with MSC-derived exosomes resulted in the uptake of exosomes by the cells, which promoted their proliferation, migration and tumorigenesis. After an extended treatment duration, the tumor cells showed morphological changes and significant changes in the expression of epithelial-mesenchymal transition (EMT) markers. Moreover, we found that FGF19 was highly expressed in MSC-exosomes and that exosomes stimulated NPC progression by activating the FGF19-FGFR4-dependent ERK signaling cascade and by modulating the EMT. All of these data indicated that exosomes participate in a novel mechanism by which MSCs influence NPC progression.
Collapse
Affiliation(s)
- Si Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Qicheng Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yunfei Xia
- Department of Rheumatology, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Bo You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Ying Shan
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Lili Bao
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Li Li
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| |
Collapse
|
44
|
Zhang X, Wang Z, Tian L, Xie J, Zou G, Jiang F. Increased Expression of FGF19 Contributes to Tumor Progression and Cell Motility of Human Thyroid Cancer. Otolaryngol Head Neck Surg 2015; 154:52-8. [PMID: 26450751 DOI: 10.1177/0194599815609534] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/11/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Numerous reports indicate a role for aberrant expression of fibroblast growth factor 19 (FGF19) in tumor development and progression, and several drugs have been developed to target it. The aim of this study was to investigate the clinical significance of FGF19 and examine whether it plays any roles in progression of thyroid cancer. STUDY DESIGN Translation research. SETTING Navy General Hospital of Chinese PLA, China. SUBJECTS AND METHODS Expression patterns of FGF19 protein in 100 paired formalin-fixed and paraffin-embedded cancerous and adjacent noncancerous tissues from patients with thyroid cancer were detected by immunohistochemistry. Then, in vitro migration and invasion assays of siRNA-targeted FGF19-transfected cells were performed. RESULTS Positive immunostaining of FGF19 protein expression was localized in cytoplasm with or without membrane of malignant cells and was observed in 82 (82.0%) of 100 patients with thyroid cancer. Statistically, the expression level of FGF19 protein in thyroid cancer tissues was significantly higher than that in normal tissues. In addition, FGF19 overexpression was significantly associated with the advanced tumor node metastasis staging (P = .008), the presence of extrathyroidal invasion (P = .01), lymph nodes metastasis (P = .01), and distant metastasis (P = .02). Furthermore, knockdown of FGF19 by transfection of siRNA-FGF19 could efficiently suppress the migration and invasion abilities of thyroid cancer cells in vitro. CONCLUSION Our data revealed that the increased expression of FGF19 might be involved in the malignant behaviors of thyroid cancer, highlighting its potential as a molecular marker for early diagnosis and as a possible target for therapeutic intervention of this disease.
Collapse
Affiliation(s)
- Xiliang Zhang
- Department of General Surgery, Navy General Hospital of Chinese PLA, Beijing, China
| | - Zhonghua Wang
- Department of General Surgery, Navy General Hospital of Chinese PLA, Beijing, China
| | - Lei Tian
- Department of General Surgery, Navy General Hospital of Chinese PLA, Beijing, China
| | - Jiangping Xie
- Department of General Surgery, Navy General Hospital of Chinese PLA, Beijing, China
| | - Guijun Zou
- Department of General Surgery, Navy General Hospital of Chinese PLA, Beijing, China
| | - Futing Jiang
- Department of General Surgery, Navy General Hospital of Chinese PLA, Beijing, China
| |
Collapse
|
45
|
Markan KR, Potthoff MJ. Metabolic fibroblast growth factors (FGFs): Mediators of energy homeostasis. Semin Cell Dev Biol 2015; 53:85-93. [PMID: 26428296 DOI: 10.1016/j.semcdb.2015.09.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/25/2015] [Indexed: 01/07/2023]
Abstract
The metabolic fibroblast growth factors (FGFs), FGF1, FGF15/19, and FGF21 differ from classic FGFs in that they modulate energy homeostasis in response to fluctuating nutrient availability. These unique mediators of metabolism regulate a number of physiological processes which contribute to their potent pharmacological properties. Administration of pharmacological doses of these FGFs causes weight loss, increases energy expenditure, and improves carbohydrate and lipid metabolism in obese animal models. However, many questions remain regarding the precise molecular and physiological mechanisms governing the effects of individual metabolic FGFs. Here we review the metabolic actions of FGF1, FGF15/19, and FGF21 while providing insights into their pharmacological effects by examining known biological functions.
Collapse
Affiliation(s)
- Kathleen R Markan
- Department of Pharmacology and University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Pharmacology and University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
46
|
Hu L, Cong L. Fibroblast growth factor 19 is correlated with an unfavorable prognosis and promotes progression by activating fibroblast growth factor receptor 4 in advanced-stage serous ovarian cancer. Oncol Rep 2015; 34:2683-91. [PMID: 26323668 DOI: 10.3892/or.2015.4212] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/22/2015] [Indexed: 11/05/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) has been confirmed to be associated with the progression and prognosis of ovarian cancer, while the underlying mechanism has not been well elucidated and the clinical significance of its ligand, fibroblast growth factor 19 (FGF19), has not been explored. To study the clinical significance of FGF19 in advanced‑stage serous ovarian cancer, we detected the expression of FGF19 and FGFR4 by immunohistochemistry (IHC), evaluated the correlation between FGF19 and clinicopathological factors by Chi-square (χ2) test, and analyzed the association between FGF19, FGFR4 and the overall survival rate using the Kaplan‑Meier method. As a result, we demonstrated that high expression of FGF19 and FGFR4 both predicted unfavorable prognosis (P=0.033 and 0.018, respectively), whereas FGF19-FGFR4 double high expression was a more sensitive prognostic factor of advanced-stage serous ovarian cancer (P<0.001). With experiments in vitro, we demonstrated that both recombinant FGF19 and secreted FGF19 promoted ovarian cancer proliferation and invasion by activating FGFR4 and the subsequent AKT-MAPK signaling pathway, suggesting that FGF19-FGFR4 signaling may auto-activate in a paracrine or autocrine manner. In conclusion, FGF19-FGFR4 double high expression was a more sensitive prognostic factor than FGF19 or FGFR4 alone in advanced-stage serous ovarian cancer. The FGF19-FGFR4 signaling pathway can promote ovarian cancer proliferation and invasion by the AKT-MAPK signaling pathway, indicating that FGF19 could be a potential therapeutic drug target of advanced-stage serous ovarian cancer.
Collapse
Affiliation(s)
- Lingling Hu
- Department of Gynecology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Lanxiang Cong
- Department of Gynecology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
47
|
Nagamatsu H, Teishima J, Goto K, Shikuma H, Kitano H, Shoji K, Inoue S, Matsubara A. FGF19 promotes progression of prostate cancer. Prostate 2015; 75:1092-101. [PMID: 25854696 DOI: 10.1002/pros.22994] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/25/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Fibroblast growth factor (FGF) signaling pathways have been reported to play important roles in prostate cancer (PCa) progression. FGF19 is one of a subfamily of FGFs that circulate in serum and act in an endocrine manner. Our objective was to investigate its role in the progression of PCa. METHODS The effect of FGF19 on the proliferation and epithelial-mesenchymal transition of LNCaP and PC3 cells was examined using MTT assay and Western blotting. Serum concentration of FGF19 was measured by ELISA in 209 patients with PCa, and the association between clinicopathological features and the presence of FGF19-positive cells in tissues derived from 155 patients who undergone radical prostatectomy was investigated. RESULTS Under androgen-deprived conditions achieved by incubation in medium with FGF19, the expression of N-cadherin in LNCaP cells was enhanced, that of E-cadherin and caspase 3 was suppressed, and the viability of LNCaP and PC3 cells was significantly enhanced. Significantly higher levels of PSA were recorded in the group determined by immunohistochemistry staining to be FGF19-positive (P = 0.0046). The 5-year biochemical recurrence-free survival rate after radical prostatectomy was 46.4% in the FGF19-positive group and 70.0% in the FGF19-negative group (P = 0.0027). In multivariate analysis, the presence of FGF19-positive tissues was an independent factor for worse prognosis after radical prostatectomy (P = 0.0052). Serum FGF19 levels in high Gleason grade group were higher than that in low Gleason grade group (P = 0.0009). CONCLUSIONS FGF19 might be associated with biochemical recurrence after radical prostatectomy by promoting cell proliferation and epithelial-mesenchymal transition of PCa.
Collapse
Affiliation(s)
- Hirotaka Nagamatsu
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Jun Teishima
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Keisuke Goto
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Shikuma
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Kitano
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koichi Shoji
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shogo Inoue
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akio Matsubara
- Department of Urology, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
48
|
Ronca R, Giacomini A, Rusnati M, Presta M. The potential of fibroblast growth factor/fibroblast growth factor receptor signaling as a therapeutic target in tumor angiogenesis. Expert Opin Ther Targets 2015; 19:1361-77. [PMID: 26125971 DOI: 10.1517/14728222.2015.1062475] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Fibroblast growth factors (FGFs) are endowed with a potent pro-angiogenic activity. Activation of the FGF/FGF receptor (FGFR) system occurs in a variety of human tumors. This may lead to neovascularization, supporting tumor progression and metastatic dissemination. Thus, a compelling biologic rationale exists for the development of anti-FGF/FGFR agents for the inhibition of tumor angiogenesis in cancer therapy. AREAS COVERED A comprehensive search on PubMed was performed to identify studies on the role of the FGF/FGFR system in angiogenesis. Endothelial FGFR signaling, the pro-angiogenic function of canonical FGFs, and their role in human tumors are described. In addition, experimental approaches aimed at the identification and characterization of nonselective and selective FGF/FGFR inhibitors and their evaluation in clinical trials are summarized. EXPERT OPINION Different approaches can be envisaged to inhibit the FGF/FGFR system, a target for the development of 'two-compartment' anti-angiogenic/anti-tumor agents, including FGFR selective and nonselective small-molecule tyrosine kinase inhibitors, anti-FGFR antibodies, and FGF ligand traps. Further studies are required to define the correlation between tumor vascularization and activation of the FGF/FGFR system and for the identification of cancer patients more likely to benefit from anti-FGF/FGFR treatments. In addition, advantages and disadvantages about the use of selective versus non-selective FGF inhibitors remain to be elucidated.
Collapse
Affiliation(s)
- Roberto Ronca
- a University of Brescia, Department of Molecular and Translational Medicine , Brescia, Italy +39 030 371 7311 ;
| | - Arianna Giacomini
- a University of Brescia, Department of Molecular and Translational Medicine , Brescia, Italy +39 030 371 7311 ;
| | - Marco Rusnati
- a University of Brescia, Department of Molecular and Translational Medicine , Brescia, Italy +39 030 371 7311 ;
| | - Marco Presta
- a University of Brescia, Department of Molecular and Translational Medicine , Brescia, Italy +39 030 371 7311 ;
| |
Collapse
|
49
|
Zhou X, Wang X. Klotho: a novel biomarker for cancer. J Cancer Res Clin Oncol 2015; 141:961-9. [PMID: 25086986 DOI: 10.1007/s00432-014-1788-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/23/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND The Klotho gene was originally identified as an anti-aging gene in 1997. Recent studies have demonstrated aberrant expression of Klotho in a number of cancers, including breast cancer, lung cancer, hepatocellular carcinoma (HCC), and so on. METHODS A literature search focusing on dysregulation of Klotho and its possible mechanisms in cancer was performed. RESULTS AND CONCLUSIONS Downregulation of Klotho was found in several cancers, such as pancreatic cancer, HCC, and other tumors. Epigenetic modulation, such as promoter methylation and histone deacetylation, also contributed to the dysregulation of Klotho in cancers. Downregulation of Klotho resulted in promoted proliferation and reduced apoptosis of cancer cells. The relevant mechanisms include the fibroblast growth factor signaling, the insulin-like growth factor 1 receptor pathway, and the Wnt/β-catenin signaling pathway. Furthermore, the Klotho protein hopefully provides new insights into cancer target treatment.
Collapse
Affiliation(s)
- Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | | |
Collapse
|
50
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1446] [Impact Index Per Article: 144.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|