1
|
Chen G, Chen L, Li X, Mohammadi M. FGF-based drug discovery: advances and challenges. Nat Rev Drug Discov 2025; 24:335-357. [PMID: 39875570 DOI: 10.1038/s41573-024-01125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
The fibroblast growth factor (FGF) family comprises 15 paracrine-acting and 3 endocrine-acting polypeptides, which govern a multitude of processes in human development, metabolism and tissue homeostasis. Therapeutic endocrine FGFs have recently advanced in clinical trials, with FGF19 and FGF21-based therapies on the cusp of approval for the treatment of primary sclerosing cholangitis and metabolic syndrome-associated steatohepatitis, respectively. By contrast, while paracrine FGFs were once thought to be promising drug candidates for wound healing, burns, tissue repair and ischaemic ailments based on their potent mitogenic and angiogenic properties, repeated failures in clinical trials have led to the widespread perception that the development of paracrine FGF-based drugs is not feasible. However, the observation that paracrine FGFs can exert FGF hormone-like metabolic activities has restored interest in these FGFs. The recent structural elucidation of the FGF cell surface signalling machinery and the formulation of a new threshold model for FGF signalling specificity have paved the way for therapeutically harnessing paracrine FGFs for the treatment of a range of metabolic diseases.
Collapse
Affiliation(s)
- Gaozhi Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingfeng Chen
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Moosa Mohammadi
- Institute of Cell Growth Factor, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Chaturantabut S, Oliver S, Frederick DT, Kim JJ, Robinson FP, Sinopoli A, Song TY, He Y, Chang YC, Rodriguez DJ, Chang L, Kesar D, Ching M, Dzvurumi R, Atari A, Tseng YY, Bardeesy N, Sellers WR. Identification of potent biparatopic antibodies targeting FGFR2 fusion-driven cholangiocarcinoma. J Clin Invest 2025; 135:e182417. [PMID: 40014401 PMCID: PMC11996885 DOI: 10.1172/jci182417] [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: 06/06/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, response rates and durability are limited due to the emergence of resistance, typically involving FGFR2 kinase domain mutations, and to suboptimal dosing, relating to drug adverse effects. Here, we develop biparatopic antibodies targeting the FGFR2 extracellular domain (ECD) as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of bivalent monospecific antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies targeting distinct epitope pairs in FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2 fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, we believe that biparatopic antibodies may serve as an innovative treatment option for patients with FGFR2-altered cholangiocarcinoma.
Collapse
MESH Headings
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/immunology
- Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/drug therapy
- Cholangiocarcinoma/immunology
- Cholangiocarcinoma/pathology
- Humans
- Mice
- Animals
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/drug therapy
- Bile Duct Neoplasms/immunology
- Bile Duct Neoplasms/pathology
- Oncogene Proteins, Fusion/immunology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Cell Line, Tumor
Collapse
Affiliation(s)
- Saireudee Chaturantabut
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Sydney Oliver
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Jiwan J. Kim
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Foxy P. Robinson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Tian-Yu Song
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Yao He
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yuan-Chen Chang
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Liang Chang
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Devishi Kesar
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Meilani Ching
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ruvimbo Dzvurumi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Adel Atari
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yuen-Yi Tseng
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nabeel Bardeesy
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - William R. Sellers
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Guyot E. Heparan sulfate chains in hepatocellular carcinoma. Gastroenterol Rep (Oxf) 2025; 13:goaf023. [PMID: 40093586 PMCID: PMC11908768 DOI: 10.1093/gastro/goaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/13/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Hepatocellular carcinoma (HCC) corresponds to the vast majority of liver cancer cases, with one of the highest mortality rates. Major advances have been made in this field both in the characterization of the molecular pathogenesis and in the development of systemic therapies. Despite these achievements, biomarkers and more efficient treatments are still needed to improve its management. Heparan sulfate (HS) chains are polysaccharides that are present at the cell surface or in the extracellular matrix that are able to bind various types of molecules, such as soluble factors, affecting their availability and thus their effects, or to contribute to interactions that position cells in their environments. Enzymes can modify HS chains after their synthesis, thus changing their properties. Numerous studies have shown HS-related proteins to be key actors that are associated with cellular effects, such as tumor growth, invasion, and metastasis, including in the context of liver carcinogenesis. The aim of this review is to provide a comprehensive overview of the biology of HS chains and their potential importance in HCC, from biological considerations to clinical development, and the identification of biomarkers, as well as therapeutic perspectives.
Collapse
Affiliation(s)
- Erwan Guyot
- Biochemistry Unit, Saint-Antoine Hospital, AP-HP Sorbonne University, Paris Cedex, France
| |
Collapse
|
4
|
Elhinnawi MA, Okita Y, Shigematsu K, Abdelaziz M, Shiratani R, Kawanishi K, Hengphasatporn K, Dang Cao TL, Shigeta Y, Kato M. GPNMB is a novel binding partner of FGFR1 that affects tumorigenic potential through AKT phosphorylation in TNBC. Cancer Sci 2025; 116:432-443. [PMID: 39609108 PMCID: PMC11786315 DOI: 10.1111/cas.16419] [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: 08/12/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer is a heterogeneous disease and is one of the most prevalent cancers in women. Triple-negative breast cancer (TNBC) is a relatively aggressive subtype of breast cancer, which is difficult to treat. Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane protein that is overexpressed in various types of cancers, including breast cancer, especially TNBC. In this study, bioinformatic analyses revealed enhanced fibroblast growth factor receptor 1 (FGFR1) signaling in patients with invasive breast cancer, and the GPNMBhigh/FGFR1high group exhibited a lower probability of relapse-free survival (RFS) than the GPNMBlow/FGFR1low group. Additionally, we observed that GPNMB and FGFR1 were essential for sphere formation, cellular migration, and epithelial-mesenchymal transition (EMT)-like changes in TNBC cells. To explore the mutual interaction between these two molecules, we conducted in silico protein-protein docking studies and molecular dynamics simulations. The results revealed that GPNMB isoform b exhibits high binding affinity for FGFR1 isoform c (FGFR1c), which correlates with cancer aggressiveness. We also confirmed the interaction between GPNMB and FGFR1 in TNBC cells. Furthermore, our study demonstrated that GPNMB is essential for AKT phosphorylation at T308 following FGF2 stimulation, resulting in high affinity for FGFR1c. Inhibition of AKT phosphorylation substantially reduces the tumorigenic potential of TNBC cells.
Collapse
Affiliation(s)
- Manar A. Elhinnawi
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
- Hormones DepartmentMedical Research and Clinical Studies Institute, National Research CentreGizaEgypt
- Stem Cells Lab Center of Excellence for Advanced SciencesNational Research CentreCairoEgypt
| | - Yukari Okita
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
- Division of Cell Dynamics, Transborder Medical Research CenterUniversity of TsukubaIbarakiJapan
| | - Katsunobu Shigematsu
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
| | - Mohammed Abdelaziz
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
- Department of Pathology, Faculty of MedicineSohag UniversitySohagEgypt
| | - Rie Shiratani
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
| | - Kunio Kawanishi
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
- Department of Anatomy, School of MedicineShowa UniversityTokyoJapan
| | | | - Thuy Linh Dang Cao
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
| | - Yasuteru Shigeta
- Center for Computational SciencesUniversity of TsukubaIbarakiJapan
| | - Mitsuyasu Kato
- Department of Experimental PathologyInstitute of Medicine, University of TsukubaIbarakiJapan
- Division of Cell Dynamics, Transborder Medical Research CenterUniversity of TsukubaIbarakiJapan
| |
Collapse
|
5
|
Nagaraj A, Srinivasa Raghavan S, Niraikulam A, Gautham N, Gunasekaran K. Sanggenol B, a plant bioactive, as a safer alternative to tackle cancer by antagonising human FGFR. J Biomol Struct Dyn 2024; 42:8331-8342. [PMID: 37551114 DOI: 10.1080/07391102.2023.2245047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Fibroblast Growth Receptor Factor (FGFR) are a family of proteins which are, in addition to their biological role, are involved in various pathological functions, such as cancer cellular proliferation, and metastasis. Deregulation of FGFRs at various points could result in malignancy. A conformational transition of the DFG (Asp-Phe-Gly) motif can switch the enzyme from a catalytically active (DFG-in) to an inactive (DFG-out) state. There are a few FDFR inhibitors which have received approval from the FDA, but these have adverse side effects. Hence, there is a demand for safer alternatives. With this aim, Ligand and Structure based virtual screening was carried to identify suitable lead molecule. In this process, Four Featured atom-based 3D Pharmacophore with quantitative structure-activity relationship analysis (3D-QSAR) was developed. The External validation of the hypothesis was carried invoking criteria such as Area under the ROC curve. Natural plant compound databases such as the Traditional Chinese medicine, NPACT and the ZINC Natural databases were chosen for pharmacophore filtering, which was followed by virtual screening against FGFR isoforms. The compound Sanggenol B was identified as the most suitable lead molecule. Structural stability of the protein-ligand complex and interactions of the ligand (Sanggenol B & the reference compound Ponatinib) with FGFR were analysed for 1000 ns (triplicate) by means of molecular simulation and the binding free energy was calculated using MMGBSA. Sanggenol B (PubChem CID: 15233694) binds effectively at the active site with favourable energies and is proposed as a safe alternative from a natural source.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Achyuta Nagaraj
- Department of Crystallography and Biophysics, University of Madras, Chennai, India
| | - Sriram Srinivasa Raghavan
- Department of Crystallography and Biophysics, University of Madras, Chennai, India
- RIKEN Centre for Computational Science, Kobe, Japan
| | - Ayyadurai Niraikulam
- Division of Biotechnology, Council of Scientific and Industrial Research-Central Leather Research Institute (CSIR-CLRI), Chennai, India
| | - Namasivayam Gautham
- Department of Crystallography and Biophysics, University of Madras, Chennai, India
| | | |
Collapse
|
6
|
Chaturantabut S, Oliver S, Frederick DT, Kim J, Robinson FP, Sinopoli A, Song TY, Rodriguez DJ, Chang L, Kesar D, He Y, Ching M, Dzvurumi R, Atari A, Tseng YY, Bardeesy N, Sellers WR. Identification of potent biparatopic antibodies targeting FGFR2 fusion driven cholangiocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613045. [PMID: 39345400 PMCID: PMC11429734 DOI: 10.1101/2024.09.16.613045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, the rate and durability of response are limited due to the emergence of resistance, typically involving acquired FGFR2 kinase domain mutations, and to sub-optimal dosing, relating to drug adverse effects. Here, we report the development of biparatopic antibodies targeting the FGFR2 extracellular domain (ECD), as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of standard bivalent monoparatopic antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies that target distinct epitope pairs along the FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2-fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, biparatopic antibodies may serve as new treatment options for patients with FGFR2-altered cholangiocarcinoma. Summary We identify biparatopic FGFR2 antibodies that are effective against FGFR2 fusion driven cholangiocarcinoma.
Collapse
|
7
|
Gędaj A, Gregorczyk P, Żukowska D, Chorążewska A, Ciura K, Kalka M, Porębska N, Opaliński Ł. Glycosylation of FGF/FGFR: An underrated sweet code regulating cellular signaling programs. Cytokine Growth Factor Rev 2024; 77:39-55. [PMID: 38719671 DOI: 10.1016/j.cytogfr.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute plasma-membrane localized signaling hubs that transmit signals from the extracellular environment to the cell interior, governing pivotal cellular processes like motility, metabolism, differentiation, division and death. FGF/FGFR signaling is critical for human body development and homeostasis; dysregulation of FGF/FGFR units is observed in numerous developmental diseases and in about 10% of human cancers. Glycosylation is a highly abundant posttranslational modification that is critical for physiological and pathological functions of the cell. Glycosylation is also very common within FGF/FGFR signaling hubs. Vast majority of FGFs (15 out of 22 members) are N-glycosylated and few FGFs are O-glycosylated. Glycosylation is even more abundant within FGFRs; all FGFRs are heavily N-glycosylated in numerous positions within their extracellular domains. A growing number of studies points on the multiple roles of glycosylation in fine-tuning FGF/FGFR signaling. Glycosylation modifies secretion of FGFs, determines their stability and affects interaction with FGFRs and co-receptors. Glycosylation of FGFRs determines their intracellular sorting, constitutes autoinhibitory mechanism within FGFRs and adjusts FGF and co-receptor recognition. Sugar chains attached to FGFs and FGFRs constitute also a form of code that is differentially decrypted by extracellular lectins, galectins, which transform FGF/FGFR signaling at multiple levels. This review focuses on the identified functions of glycosylation within FGFs and FGFRs and discusses their relevance for the cell physiology in health and disease.
Collapse
Affiliation(s)
- Aleksandra Gędaj
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Paulina Gregorczyk
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Dominika Żukowska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Aleksandra Chorążewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Krzysztof Ciura
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Marta Kalka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Natalia Porębska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| |
Collapse
|
8
|
Żukowska D, Chorążewska A, Ciura K, Gędaj A, Kalka M, Poźniak M, Porębska N, Opaliński Ł. The diverse dependence of galectin-1 and -8 on multivalency for the modulation of FGFR1 endocytosis. Cell Commun Signal 2024; 22:270. [PMID: 38750548 PMCID: PMC11094976 DOI: 10.1186/s12964-024-01661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024] Open
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is a N-glycosylated cell surface receptor tyrosine kinase, which upon recognition of specific extracellular ligands, fibroblast growth factors (FGFs), initiates an intracellular signaling. FGFR1 signaling ensures homeostasis of cells by fine-tuning essential cellular processes, like differentiation, division, motility and death. FGFR1 activity is coordinated at multiple steps and unbalanced FGFR1 signaling contributes to developmental diseases and cancers. One of the crucial control mechanisms over FGFR1 signaling is receptor endocytosis, which allows for rapid targeting of FGF-activated FGFR1 to lysosomes for degradation and the signal termination. We have recently demonstrated that N-glycans of FGFR1 are recognized by a precise set of extracellular galectins, secreted and intracellular multivalent lectins implicated in a plethora of cellular processes and altered in immune responses and cancers. Specific galectins trigger FGFR1 clustering, resulting in activation of the receptor and in initiation of intracellular signaling cascades that shape the cell physiology. Although some of galectin family members emerged recently as key players in the clathrin-independent endocytosis of specific cargoes, their impact on endocytosis of FGFR1 was largely unknown.Here we assessed the contribution of extracellular galectins to the cellular uptake of FGFR1. We demonstrate that only galectin-1 induces internalization of FGFR1, whereas the majority of galectins predominantly inhibit endocytosis of the receptor. We focused on three representative galectins: galectin-1, -7 and -8 and we demonstrate that although all these galectins directly activate FGFR1 by the receptor crosslinking mechanism, they exert different effects on FGFR1 endocytosis. Galectin-1-mediated internalization of FGFR1 doesn't require galectin-1 multivalency and occurs via clathrin-mediated endocytosis, resembling in this way the uptake of FGF/FGFR1 complex. In contrast galectin-7 and -8 impede FGFR1 endocytosis, causing stabilization of the receptor on the cell surface and prolonged propagation of the signals. Furthermore, using protein engineering approaches we demonstrate that it is possible to modulate or even fully reverse the endocytic potential of galectins.
Collapse
Affiliation(s)
- Dominika Żukowska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Aleksandra Chorążewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Krzysztof Ciura
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Aleksandra Gędaj
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Marta Kalka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Marta Poźniak
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Natalia Porębska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland.
| |
Collapse
|
9
|
Hargittay B, Mineev KS, Richter C, Sreeramulu S, Jonker HRA, Saxena K, Schwalbe H. NMR resonance assignment of a fibroblast growth factor 8 splicing isoform b. BIOMOLECULAR NMR ASSIGNMENTS 2023; 17:10.1007/s12104-023-10132-8. [PMID: 37118562 DOI: 10.1007/s12104-023-10132-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023]
Abstract
The splicing isoform b of human fibroblast growth factor 8 (FGF8b) is an important regulator of brain embryonic development. Here, we report the almost complete NMR chemical shift assignment of the backbone and aliphatic side chains of FGF8b. Obtained chemical shifts are in good agreement with the previously reported X-ray data, excluding the N-terminal gN helix, which apparently forms only in complex with the receptor. The reported data provide an NMR starting point for the investigation of FGF8b interaction with its receptors and with potential drugs or inhibitors.
Collapse
Affiliation(s)
- Bruno Hargittay
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
| | - Konstantin S Mineev
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
| | - Christian Richter
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
| | - Sridhar Sreeramulu
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
| | - Hendrik R A Jonker
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
| | - Krishna Saxena
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
- Structural Genomics Consortium, Johann Wolfgang Goethe University, Max-von-Laue-Str. 15, 60438, Frankfurt/Main, Germany
| | - Harald Schwalbe
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance (BMRZ), Johann Wolfgang Goethe University, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany.
| |
Collapse
|
10
|
Zhao X, Erhardt S, Sung K, Wang J. FGF signaling in cranial suture development and related diseases. Front Cell Dev Biol 2023; 11:1112890. [PMID: 37325554 PMCID: PMC10267317 DOI: 10.3389/fcell.2023.1112890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Suture mesenchymal stem cells (SMSCs) are a heterogeneous stem cell population with the ability to self-renew and differentiate into multiple cell lineages. The cranial suture provides a niche for SMSCs to maintain suture patency, allowing for cranial bone repair and regeneration. In addition, the cranial suture functions as an intramembranous bone growth site during craniofacial bone development. Defects in suture development have been implicated in various congenital diseases, such as sutural agenesis and craniosynostosis. However, it remains largely unknown how intricate signaling pathways orchestrate suture and SMSC function in craniofacial bone development, homeostasis, repair and diseases. Studies in patients with syndromic craniosynostosis identified fibroblast growth factor (FGF) signaling as an important signaling pathway that regulates cranial vault development. A series of in vitro and in vivo studies have since revealed the critical roles of FGF signaling in SMSCs, cranial suture and cranial skeleton development, and the pathogenesis of related diseases. Here, we summarize the characteristics of cranial sutures and SMSCs, and the important functions of the FGF signaling pathway in SMSC and cranial suture development as well as diseases caused by suture dysfunction. We also discuss emerging current and future studies of signaling regulation in SMSCs.
Collapse
Affiliation(s)
- Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, TX, United States
| | - Kihan Sung
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, TX, United States
| |
Collapse
|
11
|
Heparin is essential for optimal cell signaling by FGF21 and for regulation of βKlotho cellular stability. Proc Natl Acad Sci U S A 2023; 120:e2219128120. [PMID: 36745784 PMCID: PMC9962926 DOI: 10.1073/pnas.2219128120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
While important insights were gained about how FGF21 and other endocrine fibroblast growth factors (FGFs) bind to Klotho proteins, the exact mechanism of Klotho/FGF receptor assembly that drives receptor dimerization and activation has not been elucidated. The prevailing dogma is that Klotho proteins substitute for the loss of heparan sulfate proteoglycan (HSPG) binding to endocrine FGFs by high-affinity binding of endocrine FGF molecules to Klotho receptors. To explore a potential role of HSPG in FGF21 signaling, we have analyzed the dynamic properties of FGF21-induced FGF21-βKlotho-FGFR1c complexes on the surface of living wild-type (WT) or HSPG-deficient Chinese hamster ovary (CHO) cells by employing quantitative single-molecule fluorescence imaging analyses. Moreover, detailed analyses of FGF21 and FGF1 stimulation of cellular signaling pathways activated in WT or in HSPG-deficient CHO cells are also analyzed and compared. These experiments demonstrate that heparin is required for the formation of FGF21-βKlotho-FGFR1c complexes on the cell membrane and that binding of heparin or HSPG to FGFR1c is essential for optimal FGF21 stimulation of FGFR1c activation, mitogen-activated protein kinase responses, and intracellular Ca2+ release. It is also shown that FGF1 binding stimulates assembly of βKlotho and FGFR1c on cell membranes, resulting in endocytosis and degradation of βKlotho. We conclude that heparin or HSPG is essential for FGF21 signaling and for regulation of βKlotho cellular stability by acting as a coligand of FGFR1c.
Collapse
|
12
|
Anderson DM, Jayanthi LP, Gosavi S, Meiering EM. Engineering the kinetic stability of a β-trefoil protein by tuning its topological complexity. Front Mol Biosci 2023; 10:1021733. [PMID: 36845544 PMCID: PMC9945329 DOI: 10.3389/fmolb.2023.1021733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/02/2023] [Indexed: 02/11/2023] Open
Abstract
Kinetic stability, defined as the rate of protein unfolding, is central to determining the functional lifetime of proteins, both in nature and in wide-ranging medical and biotechnological applications. Further, high kinetic stability is generally correlated with high resistance against chemical and thermal denaturation, as well as proteolytic degradation. Despite its significance, specific mechanisms governing kinetic stability remain largely unknown, and few studies address the rational design of kinetic stability. Here, we describe a method for designing protein kinetic stability that uses protein long-range order, absolute contact order, and simulated free energy barriers of unfolding to quantitatively analyze and predict unfolding kinetics. We analyze two β-trefoil proteins: hisactophilin, a quasi-three-fold symmetric natural protein with moderate stability, and ThreeFoil, a designed three-fold symmetric protein with extremely high kinetic stability. The quantitative analysis identifies marked differences in long-range interactions across the protein hydrophobic cores that partially account for the differences in kinetic stability. Swapping the core interactions of ThreeFoil into hisactophilin increases kinetic stability with close agreement between predicted and experimentally measured unfolding rates. These results demonstrate the predictive power of readily applied measures of protein topology for altering kinetic stability and recommend core engineering as a tractable target for rationally designing kinetic stability that may be widely applicable.
Collapse
Affiliation(s)
| | - Lakshmi P. Jayanthi
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Shachi Gosavi
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Elizabeth M. Meiering
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada,*Correspondence: Elizabeth M. Meiering,
| |
Collapse
|
13
|
Park JS, Choi J, Cao L, Mohanty J, Suzuki Y, Park A, Baker D, Schlessinger J, Lee S. Isoform-specific inhibition of FGFR signaling achieved by a de-novo-designed mini-protein. Cell Rep 2022; 41:111545. [PMID: 36288716 PMCID: PMC9636537 DOI: 10.1016/j.celrep.2022.111545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 11/24/2022] Open
Abstract
Cellular signaling by fibroblast growth factor receptors (FGFRs) is a highly regulated process mediated by specific interactions between distinct subsets of fibroblast growth factor (FGF) ligands and two FGFR isoforms generated by alternative splicing: an epithelial b- and mesenchymal c-isoforms. Here, we investigate the properties of a mini-protein, mb7, developed by an in silico design strategy to bind to the ligand-binding region of FGFR2. We describe structural, biophysical, and cellular analyses demonstrating that mb7 binds with high affinity to the c-isoforms of FGFR, resulting in inhibition of cellular signaling induced by a subset of FGFs that preferentially activate c-isoforms of FGFR. Notably, as mb7 blocks interaction between FGFR with Klotho proteins, it functions as an antagonist of the metabolic hormones FGF19 and FGF21, providing mechanistic insights and strategies for the development of therapeutics for diseases driven by aberrantly activated FGFRs. Park et al. show that a de-novo-designed mini-protein, mb7, can specifically recognize c-isoforms of FGFRs. By masking the regions of FGFR that are critical for the FGFR activation, mb7 can potently inhibit cellular signaling by a subset of FGFs that preferentially activate FGFR c-isoform signaling.
Collapse
|
14
|
Wu X, Liu Z, Gan C, Wei W, Zhang Q, Liu H, Que H, Su X, Yue L, He H, Ouyang L, Ye T. Design, synthesis and biological evaluation of a series of novel pyrrolo[2,3-d]pyrimidin/pyrazolo[3,4-d]pyrimidin-4-amine derivatives as FGFRs-dominant multi-target receptor tyrosine kinase inhibitors for the treatment of gastric cancer. Bioorg Chem 2022; 127:105965. [PMID: 35759882 DOI: 10.1016/j.bioorg.2022.105965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/08/2022] [Accepted: 06/10/2022] [Indexed: 02/05/2023]
Abstract
Gastric cancer is the second most lethal cancer across the world. With the progress in therapeutic approaches, the 5-year survival rate of early gastric cancer can reach > 95%. However, the prognosis and survival time of advanced gastric cancer is still somber. Therefore, more effective targeted therapies for gastric cancer treatment are urgently needed. FGFR, VEGFR and other receptor tyrosine kinases have recently been suggested as potential targets for gastric cancer treatment. We herein report the discovery of pyrrolo[2,3-d]pyrimidin/pyrazolo[3,4-d]pyrimidin-4-amine derivatives as a new class of FGFRs-dominant multi-target receptor tyrosine kinase inhibitors. SAR assessment identified the most active compounds 8f and 8k, which showed excellent inhibitory activity against a variety of receptor tyrosine kinases. Moreover, 8f and 8k displayed excellent potency in the SNU-16 gastric cancer cell line. Furthermore, 8f and 8k could inhibit FGFR1 phosphorylation and downstream signaling pathways as well as induce cell apoptosis. In vivo, 8f and 8k suppress tumor growth in the SNU-16 xenograft model without inducing obvious toxicity. These findings raise the possibility that compounds 8f and 8k might serve as potential agents for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Xiuli Wu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihao Liu
- Laboratory of Emergency Medicine, Department of Emergency Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cailin Gan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Wei
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qianyu Zhang
- West China School of Public Health and Healthy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyao Liu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hanyun Que
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingping Su
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Yue
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hualong He
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Ouyang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tinghong Ye
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
15
|
Anti-tumor and anti-metastatic activity of the FGF2 118-126 fragment dependent on the loop structure. Biochem J 2022; 479:1285-1302. [PMID: 35638868 DOI: 10.1042/bcj20210830] [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: 12/09/2021] [Revised: 05/20/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Fibroblast Growth Factor /FGF Receptor 1 (FGF2/FGFR1) system regulates the growth and metastasis of different cancers. Inhibition of this signaling pathway is an attractive target for cancer therapy. Here, we aimed to reproduce the 118-126 fragment of FGF2 to interfere with the FGF2-FGFR1 interaction. To determine whether the loop structure affects the function of this fragment, we compared cyclic (disulfide-bonded) and linear peptide variants. The cyclic peptide (referred to as BGF1) effectively inhibited the FGF2-induced proliferation of HUVECs, 4T1 mammary carcinoma, U87 glioblastoma, and SKOV3 ovarian carcinoma cells. It led to apoptosis induction in HUVECs, whereas the linear peptide (referred to as BGF2) was ineffective. In a murine 4T1 tumor model, BGF1 inhibited tumor growth more effectively than Avastin and increased animals' survival without causing weight loss, but the linear peptide BGF2 had no significant anti-tumor effects. According to immunohistochemical studies, the anti-tumor properties of BGF1 were associated with suppression of tumor cell proliferation (Ki-67 expression), angiogenesis (CD31 expression), and apoptosis induction (as was shown by increased p53 expression and TUNEL staining and decreased Bcl-2 expression). The potential of BGF1 to suppress tumor invasion was indicated by quantitative analysis of the metastasis-related proteins, including FGFR1, pFGFR1, NF-κB, p-NF-κB, MMP-9, E-cadherin, N-cadherin, and Vimentin, and supported by small animal positron emission tomography (PET) used 18Fluorodeoxyglucose (18F-FDG). These results demonstrate that the functional properties of the 118-126 region of FGF2 depend on the loop structure and the peptide derived from this fragment encourages further preclinical investigations.
Collapse
|
16
|
Blaber M. Variable and Conserved Regions of Secondary Structure in the β-Trefoil Fold: Structure Versus Function. Front Mol Biosci 2022; 9:889943. [PMID: 35517858 PMCID: PMC9062101 DOI: 10.3389/fmolb.2022.889943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
β-trefoil proteins exhibit an approximate C3 rotational symmetry. An analysis of the secondary structure for members of this diverse superfamily of proteins indicates that it is comprised of remarkably conserved β-strands and highly-divergent turn regions. A fundamental “minimal” architecture can be identified that is devoid of heterogenous and extended turn regions, and is conserved among all family members. Conversely, the different functional families of β-trefoils can potentially be identified by their unique turn patterns (or turn “signature”). Such analyses provide clues as to the evolution of the β-trefoil family, suggesting a folding/stability role for the β-strands and a functional role for turn regions. This viewpoint can also guide de novo protein design of β-trefoil proteins having novel functionality.
Collapse
Affiliation(s)
- Michael Blaber
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
17
|
Mohale M, Gundampati RK, Krishnaswamy Suresh Kumar T, Heyes CD. Site-specific labeling and functional efficiencies of human fibroblast growth Factor-1 with a range of fluorescent Dyes in the flexible N-Terminal region and a rigid β-turn region. Anal Biochem 2022; 640:114524. [PMID: 34933004 DOI: 10.1016/j.ab.2021.114524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/08/2021] [Indexed: 11/01/2022]
Abstract
Human fibroblast growth factor-1 (hFGF1) binding to its receptor and heparin play critical roles in cell proliferation, angiogenesis and wound healing but is also implicated in cancer. Fluorescence imaging is a powerful approach to study such protein interactions, but it is not always obvious if the site chosen will be efficiently labeled, often relying on trial-and-error. To provide a more systematic approach towards an efficient site-specific labeling strategy, we labeled two structurally distinct regions of the protein - the flexible N-terminus and a rigid loop. Several dyes were chosen to cover the visible region and to investigate how the structure of the dye affects the labeling efficiency. Flexibility in either the protein labeling site or the dye structure was found to result in high labeling efficiency, but flexibility in both resulted in a significant decrease in labeling efficiency. Conversely, too much rigidity in both can result in dye-protein interactions that can aggregate the protein. Importantly, site-specifically labeling hFGF1 in these regions maintained biological activity. These results could be applicable to other proteins by considering the flexibility of both the protein labeling site and the dye structure.
Collapse
Affiliation(s)
- Mamello Mohale
- Department of Chemistry and Biochemistry, University of Arkansas, 345 N. Campus Drive, Fayetteville, AR, 72701, USA
| | - Ravi Kumar Gundampati
- Department of Chemistry and Biochemistry, University of Arkansas, 345 N. Campus Drive, Fayetteville, AR, 72701, USA
| | | | - Colin D Heyes
- Department of Chemistry and Biochemistry, University of Arkansas, 345 N. Campus Drive, Fayetteville, AR, 72701, USA.
| |
Collapse
|
18
|
Chen L, Zhang Y, Yin L, Cai B, Huang P, Li X, Liang G. Fibroblast growth factor receptor fusions in cancer: opportunities and challenges. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:345. [PMID: 34732230 PMCID: PMC8564965 DOI: 10.1186/s13046-021-02156-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022]
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) play critical roles in many biological processes and developmental functions. Chromosomal translocation of FGFRs result in the formation of chimeric FGFR fusion proteins, which often cause aberrant signaling leading to the development and progression of human cancer. Due to the high recurrence rate and carcinogenicity, oncogenic FGFR gene fusions have been identified as promising therapeutic targets. Erdafitinib and pemigatinib, two FGFR selective inhibitors targeting FGFR fusions, have been approved by the U.S. Food and Drug Administration (FDA) to treat patients with urothelial cancer and cholangiocarcinoma, respectively. Futibatinib, a third-generation FGFR inhibitor, is under phase III clinical trials in patients with FGFR gene rearrangements. Herein, we review the current understanding of the FGF/FGFRs system and the oncogenic effect of FGFR fusions, summarize promising inhibitors under clinical development for patients with FGFR fusions, and highlight the challenges in this field.
Collapse
Affiliation(s)
- Lingfeng Chen
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China. .,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310012, Zhejiang, China.
| | - Yanmei Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310012, Zhejiang, China
| | - Lina Yin
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310012, Zhejiang, China
| | - Binhao Cai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Guang Liang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China. .,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310012, Zhejiang, China. .,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
19
|
Cheong DE, Choi HJ, Yoo SK, Lee HD, Kim GJ. A designed fusion tag for soluble expression and selective separation of extracellular domains of fibroblast growth factor receptors. Sci Rep 2021; 11:21453. [PMID: 34728710 PMCID: PMC8563715 DOI: 10.1038/s41598-021-01029-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/18/2021] [Indexed: 11/10/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) generate various transduction signals by interaction with fibroblast growth factors (FGFs) and are involved in various biological functions such as cell proliferation, migration, and differentiation. Malfunction of these proteins may lead to the development of various diseases, including cancer. Accordingly, FGFRs are considered an alternative therapeutic target for protein and/or gene therapy. However, the screening of antagonists or agonists of FGFRs is challenging due to their complex structural features associated with protein expression. Herein, we conducted the development of a protease-free cleavable tag (PFCT) for enhancing the solubility of difficult-to express protein by combining maltose-binding protein (MBP) and the C-terminal region of Npu intein. To validate the availability of the resulting tag for the functional production of extracellular domains of FGFRs (Ec_FGFRs), we performed fusion of PFCT with the N-terminus of Ec_FGFRs and analyzed the expression patterns. Almost all PFCT-Ec_FGFR fusion proteins were mainly detected in the soluble fraction except for Ec_FGFR4. Upon addition of the N-terminal region of Npu intein, approximately 85% of the PFCT-Ec_FGFRs was separated into PFCT and Ec_FGFR via intein-mediated cleavage. Additionally, the structural integrity of Ec_FGFR was confirmed by affinity purification using heparin column. Taken together, our study demonstrated that the PFCT could be used for soluble expression and selective separation of Ec_FGFRs.
Collapse
Affiliation(s)
- Dae-Eun Cheong
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Sciences, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, 61186, Korea
| | - Hye-Ji Choi
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Sciences, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, 61186, Korea
| | - Su-Kyoung Yoo
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Sciences, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, 61186, Korea
| | - Hun-Dong Lee
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Sciences, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, 61186, Korea
| | - Geun-Joong Kim
- Department of Biological Sciences and Research Center of Ecomimetics, College of Natural Sciences, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, 61186, Korea.
| |
Collapse
|
20
|
Pozo F, Martinez-Gomez L, Walsh TA, Rodriguez JM, Di Domenico T, Abascal F, Vazquez J, Tress ML. Assessing the functional relevance of splice isoforms. NAR Genom Bioinform 2021; 3:lqab044. [PMID: 34046593 PMCID: PMC8140736 DOI: 10.1093/nargab/lqab044] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022] Open
Abstract
Alternative splicing of messenger RNA can generate an array of mature transcripts, but it is not clear how many go on to produce functionally relevant protein isoforms. There is only limited evidence for alternative proteins in proteomics analyses and data from population genetic variation studies indicate that most alternative exons are evolving neutrally. Determining which transcripts produce biologically important isoforms is key to understanding isoform function and to interpreting the real impact of somatic mutations and germline variations. Here we have developed a method, TRIFID, to classify the functional importance of splice isoforms. TRIFID was trained on isoforms detected in large-scale proteomics analyses and distinguishes these biologically important splice isoforms with high confidence. Isoforms predicted as functionally important by the algorithm had measurable cross species conservation and significantly fewer broken functional domains. Additionally, exons that code for these functionally important protein isoforms are under purifying selection, while exons from low scoring transcripts largely appear to be evolving neutrally. TRIFID has been developed for the human genome, but it could in principle be applied to other well-annotated species. We believe that this method will generate valuable insights into the cellular importance of alternative splicing.
Collapse
Affiliation(s)
- Fernando Pozo
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Martinez-Gomez
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Thomas A Walsh
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - José Manuel Rodriguez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Tomas Di Domenico
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Federico Abascal
- Somatic Evolution Group, Wellcome Sanger Institute, Hinxton CB10 1SA, UK
| | - Jesús Vazquez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Michael L Tress
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
21
|
Mohan S, Muthusamy K, Nagamani S, Kesavan C. Computational prediction of small molecules with predicted binding to FGFR3 and testing biological effects in bone cells. Exp Biol Med (Maywood) 2021; 246:1660-1667. [PMID: 33779341 DOI: 10.1177/15353702211002181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Activating anabolic receptor-mediated signaling is essential for stimulating new bone formation and for promoting bone healing in humans. Fibroblast growth factor receptor (FGFR) 3 is reported to be an important positive regulator of osteogenesis. Presently, recombinant proteins are used to stimulate FGFR3 function but have limitations for therapy due to expense and stability. Therefore, there is a need for identification of novel small molecules binding to FGFR3 that promote biological function. In silico molecular docking and high-throughput virtual screening on zinc database identified seven compounds predicted to bind to an active site within the βC'-βE loop, specific to FGFR3. All seven compounds fall within an acceptable range of ADME/T properties. Four compounds showed a 30-65% oral absorption rate. Density functional theory analysis revealed a high HOMO-LUMO gap, reflecting high molecular stability for compounds 14977614 and 13509082. Five compounds exhibited mutagenicity, while the other three compounds presented irritability. Computational mutagenesis predicted that mutating G322 affected compound binding to FGFR3. Molecular dynamics simulation revealed compound 14977614 is stable in binding to FGFR3. Furthermore, compound 14977614, with an oral absorption rate of 60% and high molecular stability, produced significant increases in both proliferation and differentiation of bone marrow stromal cells in vitro. Anti-FGFR3 treatment completely blocked the stimulatory effect of 14977614 on BMSC proliferation. Ex vivo treatment of mouse calvaria in organ culture for seven days with 14977614 increased mineralization and expression levels of bone formation markers. In conclusion, computational analyses identified seven compounds that bind to the FGFR3, and in vitro studies showed that compound 14977614 exerts significant biological effects on osteogenic cells.
Collapse
Affiliation(s)
- Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA.,Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA.,Department of Orthopedic Surgery, Loma Linda University, Loma Linda, CA 92354, USA
| | | | | | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA.,Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
22
|
FGF23 contains two distinct high-affinity binding sites enabling bivalent interactions with α-Klotho. Proc Natl Acad Sci U S A 2020; 117:31800-31807. [PMID: 33257569 DOI: 10.1073/pnas.2018554117] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The three members of the endocrine-fibroblast growth factor (FGF) family, FGF19, 21, and 23 are circulating hormones that regulate critical metabolic processes. FGF23 stimulates the assembly of a signaling complex composed of α-Klotho (KLA) and FGF receptor (FGFR) resulting in kinase activation, regulation of phosphate homeostasis, and vitamin D levels. Here we report that the C-terminal tail of FGF23, a region responsible for KLA binding, contains two tandem repeats, repeat 1 (R1) and repeat 2 (R2) that function as two distinct ligands for KLA. FGF23 variants with a single KLA binding site, FGF23-R1, FGF23-R2, or FGF23-wild type (WT) with both R1 and R2, bind to KLA with similar binding affinity and stimulate FGFR1 activation and MAPK response. R2 is flanked by two cysteines that form a disulfide bridge in FGF23-WT; disulfide bridge formation in FGF23-WT is dispensable for KLA binding and for cell signaling via FGFRs. We show that FGF23-WT stimulates dimerization and activation of a chimeric receptor molecule composed of the extracellular domain of KLA fused to the cytoplasmic domain of FGFR and employ total internal reflection fluorescence microscopy to visualize individual KLA molecules on the cell surface. These experiments demonstrate that FGF23-WT can act as a bivalent ligand of KLA in the cell membrane. Finally, an engineered Fc-R2 protein acts as an FGF23 antagonist offering new pharmacological intervention for treating diseases caused by excessive FGF23 abundance or activity.
Collapse
|
23
|
Schittenhelm J, Ziegler L, Sperveslage J, Mittelbronn M, Capper D, Burghardt I, Poso A, Biskup S, Skardelly M, Tabatabai G. FGFR3 overexpression is a useful detection tool for FGFR3 fusions and sequence variations in glioma. Neurooncol Pract 2020; 8:209-221. [PMID: 33898054 DOI: 10.1093/nop/npaa075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Fibroblast growth factor receptor (FGFR) inhibitors are currently used in clinical development. A subset of glioblastomas carries gene fusion of FGFR3 and transforming acidic coiled-coil protein 3. The prevalence of other FGFR3 alterations in glioma is currently unclear. Methods We performed RT-PCR in 101 glioblastoma samples to detect FGFR3-TACC3 fusions ("RT-PCR cohort") and correlated results with FGFR3 immunohistochemistry (IHC). Further, we applied FGFR3 IHC in 552 tissue microarray glioma samples ("TMA cohort") and validated these results in two external cohorts with 319 patients. Gene panel sequencing was carried out in 88 samples ("NGS cohort") to identify other possible FGFR3 alterations. Molecular modeling was performed on newly detected mutations. Results In the "RT-PCR cohort," we identified FGFR3-TACC3 fusions in 2/101 glioblastomas. Positive IHC staining was observed in 73/1024 tumor samples of which 10 were strongly positive. In the "NGS cohort," we identified FGFR3 fusions in 9/88 cases, FGFR3 amplification in 2/88 cases, and FGFR3 gene mutations in 7/88 cases in targeted sequencing. All FGFR3 fusions and amplifications and a novel FGFR3 K649R missense mutation were associated with FGFR3 overexpression (sensitivity and specificity of 93% and 95%, respectively, at cutoff IHC score > 7). Modeling of these data indicated that Tyr647, a residue phosphorylated as a part of FGFR3 activation, is affected by the K649R mutation. Conclusions FGFR3 IHC is a useful screening tool for the detection of FGFR3 alterations and could be included in the workflow for isocitrate dehydrogenase (IDH) wild-type glioma diagnostics. Samples with positive FGFR3 staining could then be selected for NGS-based diagnostic tools.
Collapse
Affiliation(s)
- Jens Schittenhelm
- Center for Neuro-Oncology, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.,Department of Neuropathology, Institute of Pathology and Neuropathology, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Lukas Ziegler
- Department of Neuropathology, Institute of Pathology and Neuropathology, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Jan Sperveslage
- Department of Pathology, Institute of Pathology and Neuropathology, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.,Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Michel Mittelbronn
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg.,Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg.,Department of Oncology (DONC), Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg.,Edinger Institute (Neurological Institute), University of Frankfurt, Frankfurt, Germany
| | - David Capper
- Institute of Neuropathology, Berlin Institute of Health, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Isabel Burghardt
- Center for Neuro-Oncology, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.,Department of Neurology & Interdisciplinary Neurooncology, University Hospital Tübingen, Hertie-Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tuebingen, Germany
| | - Antti Poso
- Department of Internal Medicine VIII, University Hospital Tuebingen, Tuebingen, Germany
| | - Saskia Biskup
- CeGaT GmbH and Praxis für Humangenetik Tuebingen, Tuebingen, Germany
| | - Marco Skardelly
- Center for Neuro-Oncology, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.,Department of Neurosurgery, University Hospital of Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ghazaleh Tabatabai
- Center for Neuro-Oncology, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.,Department of Neurology & Interdisciplinary Neurooncology, University Hospital Tübingen, Hertie-Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tuebingen, Germany.,Center for Personalized Medicine, Eberhard Karls University of Tuebingen, Tuebingen, Germany.,German Consortium for Translational Cancer Research (DKTK), DKFZ partner site Tuebingen, Tuebingen, Germany
| |
Collapse
|
24
|
Parico GCG, Partch CL. The tail of cryptochromes: an intrinsically disordered cog within the mammalian circadian clock. Cell Commun Signal 2020; 18:182. [PMID: 33198762 PMCID: PMC7667820 DOI: 10.1186/s12964-020-00665-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022] Open
Abstract
Cryptochrome (CRY) proteins play an essential role in regulating mammalian circadian rhythms. CRY is composed of a structured N-terminal domain known as the photolyase homology region (PHR), which is tethered to an intrinsically disordered C-terminal tail. The PHR domain is a critical hub for binding other circadian clock components such as CLOCK, BMAL1, PERIOD, or the ubiquitin ligases FBXL3 and FBXL21. While the isolated PHR domain is necessary and sufficient to generate circadian rhythms, removing or modifying the cryptochrome tails modulates the amplitude and/or periodicity of circadian rhythms, suggesting that they play important regulatory roles in the molecular circadian clock. In this commentary, we will discuss how recent studies of these intrinsically disordered tails are helping to establish a general and evolutionarily conserved model for CRY function, where the function of PHR domains is modulated by reversible interactions with their intrinsically disordered tails. Video abstract
Collapse
Affiliation(s)
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, UC Santa Cruz, Santa Cruz, USA. .,Center for Circadian Biology, UC San Diego, La Jolla, USA.
| |
Collapse
|
25
|
Parico GCG, Perez I, Fribourgh JL, Hernandez BN, Lee HW, Partch CL. The human CRY1 tail controls circadian timing by regulating its association with CLOCK:BMAL1. Proc Natl Acad Sci U S A 2020; 117:27971-27979. [PMID: 33106415 PMCID: PMC7668087 DOI: 10.1073/pnas.1920653117] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Circadian rhythms are generated by interlocked transcription-translation feedback loops that establish cell-autonomous biological timing of ∼24 h. Mutations in core clock genes that alter their stability or affinity for one another lead to changes in circadian period. The human CRY1Δ11 mutant lengthens circadian period to cause delayed sleep phase disorder (DSPD), characterized by a very late onset of sleep. CRY1 is a repressor that binds to the transcription factor CLOCK:BMAL1 to inhibit its activity and close the core feedback loop. We previously showed how the PHR (photolyase homology region) domain of CRY1 interacts with distinct sites on CLOCK and BMAL1 to sequester the transactivation domain from coactivators. However, the Δ11 variant alters an intrinsically disordered tail in CRY1 downstream of the PHR. We show here that the CRY1 tail, and in particular the region encoded by exon 11, modulates the affinity of the PHR domain for CLOCK:BMAL1. The PHR-binding epitope in exon 11 is necessary and sufficient to disrupt the interaction between CRY1 and the subunit CLOCK. Moreover, PHR-tail interactions are conserved in the paralog CRY2 and reduced when either CRY is bound to the circadian corepressor PERIOD2. Discovery of this autoregulatory role for the mammalian CRY1 tail and conservation of PHR-tail interactions in both mammalian cryptochromes highlights functional conservation with plant and insect cryptochromes, which also utilize PHR-tail interactions to reversibly control their activity.
Collapse
Affiliation(s)
- Gian Carlo G Parico
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064
| | - Ivette Perez
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064
| | - Jennifer L Fribourgh
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064
| | - Britney N Hernandez
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064
| | - Hsiau-Wei Lee
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064;
- Center for Circadian Biology, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
26
|
Jin W, Jiang D, Zhang W, Wang C, Xia K, Zhang F, Linhardt RJ. Interactions of fibroblast growth factors with sulfated galactofucan from Saccharina japonica. Int J Biol Macromol 2020; 160:26-34. [PMID: 32464202 PMCID: PMC10466213 DOI: 10.1016/j.ijbiomac.2020.05.183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/14/2020] [Accepted: 05/22/2020] [Indexed: 01/09/2023]
Abstract
A total 68 types of marine algae oligosaccharides and polysaccharides were prepared and used to study the structure-activity relationship of oligosaccharides and polysaccharides in their interactions with fibroblast growth factors (FGF) 1 and 2. Factors considered include different types of algae, extraction methods, molecular weight, sulfate content and fractions. In the case of low molecular weight polysaccharide (SJ-D) from Saccharina japonica and its fractions eluting from anion exchange column, both 1.0 M NaCl fraction (SJ-D-I) and 2.0 M NaCl fraction (SJ-D-S) had stronger binding affinity than the parent SJ-D, suggesting that sulfated galactofucans represented the major tight binding component. Nuclear magnetic resonance showed that SJ-D-I was a typical sulfated galactofucan, composed of four units: 1, 3-linked 4-sulfated α-L-fucose (Fuc); 1, 3-linked 2, 4-disulfated α-L-Fuc; 1, 6-linked 4-sulfated β-D-Gal and/or 1, 6-linked 3, 4-sulfated β-D-Gal. Modification by autohydrolysis to oligosaccharides and desulfation decreased the FGF binding affinity while oversulfation increased the affinity. The solution-based affinities of SJ-D-I to FGF1 and FGF2 were 69 nM and 3.9 nM, suggesting that SJ-D-I showed better preferentially binding to FGF1 than a natural ligand, heparin, suggesting that sulfated galactofucan might represent a good regulator of FGF1.
Collapse
Affiliation(s)
- Weihua Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China; Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Di Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wenjing Zhang
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Chunyu Wang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA; Department of Biological Science, Departments of Chemistry and Chemical Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Ke Xia
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA.
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA; Department of Biological Science, Departments of Chemistry and Chemical Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| |
Collapse
|
27
|
Levine KM, Ding K, Chen L, Oesterreich S. FGFR4: A promising therapeutic target for breast cancer and other solid tumors. Pharmacol Ther 2020; 214:107590. [PMID: 32492514 PMCID: PMC7494643 DOI: 10.1016/j.pharmthera.2020.107590] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
The fibroblast growth factor receptor (FGFR) signaling pathway has long been known to cancer researchers because of its role in cell survival, proliferation, migration, and angiogenesis. Dysregulation of FGFR signaling is frequently reported in cancer studies, but most of these studies focus on FGFR1-3. However, there is growing evidence implicating an important and unique role of FGFR4 in oncogenesis, tumor progression, and resistance to anti-tumor therapy in multiple types of cancer. Importantly, there are several novel FGFR4-specific inhibitors in clinical trials, making FGFR4 an attractive target for further research. In this review, we focus on assessing the role of FGFR4 in cancer, with an emphasis on breast cancer. First, the structure, physiological functions and downstream signaling pathways of FGFR4 are introduced. Next, different mechanisms reported to cause aberrant FGFR4 activation and their functions in cancer are discussed, including FGFR4 overexpression, FGF ligand overexpression, FGFR4 somatic hotspot mutations, and the FGFR4 G388R single nucleotide polymorphism. Finally, ongoing and recently completed clinical trials targeting FGFRs in cancer are reviewed, highlighting the therapeutic potential of FGFR4 inhibition for the treatment of breast cancer.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Gene Expression Regulation, Neoplastic
- Molecular Targeted Therapy
- Mutation
- Polymorphism, Single Nucleotide
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Kevin M Levine
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kai Ding
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lyuqin Chen
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Magee-Women's Research Institute, Magee-Women's Research Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol 2020; 16:547-564. [PMID: 32807927 DOI: 10.1038/s41584-020-0469-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Regulated fibroblast growth factor (FGF) signalling is a prerequisite for the correct development and homeostasis of articular cartilage, as evidenced by the fact that aberrant FGF signalling contributes to the maldevelopment of joints and to the onset and progression of osteoarthritis. Of the four FGF receptors (FGFRs 1-4), FGFR1 and FGFR3 are strongly implicated in osteoarthritis, and FGFR1 antagonists, as well as agonists of FGFR3, have shown therapeutic efficacy in mouse models of spontaneous and surgically induced osteoarthritis. FGF18, a high affinity ligand for FGFR3, is the only FGF-based drug currently in clinical trials for osteoarthritis. This Review covers the latest advances in our understanding of the molecular mechanisms that regulate FGF signalling during normal joint development and in the pathogenesis of osteoarthritis. Strategies for FGF signalling-based treatment of osteoarthritis and for cartilage repair in animal models and clinical trials are also introduced. An improved understanding of FGF signalling from a structural biology perspective, and of its roles in skeletal development and diseases, could unlock new avenues for discovery of modulators of FGF signalling that can slow or stop the progression of osteoarthritis.
Collapse
|
29
|
Matsuoka D, Kamiya M, Sato T, Sugita Y. Role of the N-Terminal Transmembrane Helix Contacts in the Activation of FGFR3. J Comput Chem 2019; 41:561-572. [PMID: 31804721 DOI: 10.1002/jcc.26122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
Abstract
Fibroblast growth factor receptor 3 (FGFR3) is a member of receptor tyrosine kinases, which is involved in skeletal cell growth, differentiation, and migration. FGFR3 transduces biochemical signals from the extracellular ligand-binding domain to the intracellular kinase domain through the conformational changes of the transmembrane (TM) helix dimer. Here, we apply generalized replica exchange with solute tempering method to wild type (WT) and G380R mutant (G380R) of FGFR3. The dimer interface in G380R is different from WT and the simulation results are in good agreement with the solid-state nuclear magnetic resonance (NMR) spectroscopy. TM helices in G380R are extended more than WT, and thereby, G375 in G380R contacts near the N-termini of the TM helix dimer. Considering that both G380R and G375C show the constitutive activation, the formation of the N-terminal contacts of the TM helices can be generally important for the activation mechanism. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daisuke Matsuoka
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, 351-0198, Japan
| | - Motoshi Kamiya
- Computational Biophysics Research Team, RIKEN Center for Computational Science, Kobe, 650-0047, Japan
| | - Takeshi Sato
- Division of Liberal Arts and Science, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Yuji Sugita
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, 351-0198, Japan.,Computational Biophysics Research Team, RIKEN Center for Computational Science, Kobe, 650-0047, Japan.,Laboratory for Biomolecular Function Simulation, RIKEN Center for Biosystem Dynamics Research, Kobe, 650-0047, Japan
| |
Collapse
|
30
|
Jimenez-Pascual A, Siebzehnrubl FA. Fibroblast Growth Factor Receptor Functions in Glioblastoma. Cells 2019; 8:E715. [PMID: 31337028 PMCID: PMC6678715 DOI: 10.3390/cells8070715] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is the most lethal brain cancer in adults, with no known cure. This cancer is characterized by a pronounced genetic heterogeneity, but aberrant activation of receptor tyrosine kinase signaling is among the most frequent molecular alterations in glioblastoma. Somatic mutations of fibroblast growth factor receptors (FGFRs) are rare in these cancers, but many studies have documented that signaling through FGFRs impacts glioblastoma progression and patient survival. Small-molecule inhibitors of FGFR tyrosine kinases are currently being trialed, underlining the therapeutic potential of blocking this signaling pathway. Nevertheless, a comprehensive overview of the state of the art of the literature on FGFRs in glioblastoma is lacking. Here, we review the evidence for the biological functions of FGFRs in glioblastoma, as well as pharmacological approaches to targeting these receptors.
Collapse
MESH Headings
- Brain Neoplasms/metabolism
- Disease Progression
- Glioblastoma/metabolism
- Humans
- Receptor, Fibroblast Growth Factor, Type 1/chemistry
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- Receptor, Fibroblast Growth Factor, Type 2/chemistry
- Receptor, Fibroblast Growth Factor, Type 2/physiology
- Receptor, Fibroblast Growth Factor, Type 3/chemistry
- Receptor, Fibroblast Growth Factor, Type 3/physiology
- Receptor, Fibroblast Growth Factor, Type 4/chemistry
- Receptor, Fibroblast Growth Factor, Type 4/physiology
Collapse
Affiliation(s)
- Ana Jimenez-Pascual
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK.
| |
Collapse
|
31
|
Dai S, Zhou Z, Chen Z, Xu G, Chen Y. Fibroblast Growth Factor Receptors (FGFRs): Structures and Small Molecule Inhibitors. Cells 2019; 8:E614. [PMID: 31216761 PMCID: PMC6627960 DOI: 10.3390/cells8060614] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/05/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases expressed on the cell membrane that play crucial roles in both developmental and adult cells. Dysregulation of FGFRs has been implicated in a wide variety of cancers, such as urothelial carcinoma, hepatocellular carcinoma, ovarian cancer and lung adenocarcinoma. Due to their functional importance, FGFRs have been considered as promising drug targets for the therapy of various cancers. Multiple small molecule inhibitors targeting this family of kinases have been developed, and some of them are in clinical trials. Furthermore, the pan-FGFR inhibitor erdafitinib (JNJ-42756493) has recently been approved by the U.S. Food and Drug Administration (FDA) for the treatment of metastatic or unresectable urothelial carcinoma (mUC). This review summarizes the structure of FGFR, especially its kinase domain, and the development of small molecule FGFR inhibitors.
Collapse
Affiliation(s)
- Shuyan Dai
- NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Zhan Zhou
- NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Zhuchu Chen
- NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Guangyu Xu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Yongheng Chen
- NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
32
|
Cross-Talk between Fibroblast Growth Factor Receptors and Other Cell Surface Proteins. Cells 2019; 8:cells8050455. [PMID: 31091809 PMCID: PMC6562592 DOI: 10.3390/cells8050455] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute signaling circuits that transmit signals across the plasma membrane, regulating pivotal cellular processes like differentiation, migration, proliferation, and apoptosis. The malfunction of FGFs/FGFRs signaling axis is observed in numerous developmental and metabolic disorders, and in various tumors. The large diversity of FGFs/FGFRs functions is attributed to a great complexity in the regulation of FGFs/FGFRs-dependent signaling cascades. The function of FGFRs is modulated at several levels, including gene expression, alternative splicing, posttranslational modifications, and protein trafficking. One of the emerging ways to adjust FGFRs activity is through formation of complexes with other integral proteins of the cell membrane. These proteins may act as coreceptors, modulating binding of FGFs to FGFRs and defining specificity of elicited cellular response. FGFRs may interact with other cell surface receptors, like G-protein-coupled receptors (GPCRs) or receptor tyrosine kinases (RTKs). The cross-talk between various receptors modulates the strength and specificity of intracellular signaling and cell fate. At the cell surface FGFRs can assemble into large complexes involving various cell adhesion molecules (CAMs). The interplay between FGFRs and CAMs affects cell–cell interaction and motility and is especially important for development of the central nervous system. This review summarizes current stage of knowledge about the regulation of FGFRs by the plasma membrane-embedded partner proteins and highlights the importance of FGFRs-containing membrane complexes in pathological conditions, including cancer.
Collapse
|
33
|
Zinkle A, Mohammadi M. Structural Biology of the FGF7 Subfamily. Front Genet 2019; 10:102. [PMID: 30809251 PMCID: PMC6379346 DOI: 10.3389/fgene.2019.00102] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/29/2019] [Indexed: 11/13/2022] Open
Abstract
Mammalian fibroblast growth factor (FGF) signaling is intricately regulated via selective binding interactions between 18 FGF ligands and four FGF receptors (FGFR1–4), three of which (FGFR1–3) are expressed as either epithelial (“b”) or mesenchymal (“c”) splice isoforms. The FGF7 subfamily, consisting of FGF3, FGF7, FGF10, and FGF22, is unique among FGFs in that its members are secreted exclusively by the mesenchyme, and specifically activate the “b” isoforms of FGFR1 (FGFR1b) and FGFR2 (FGFR2b) present in the overlying epithelium. This unidirectional mesenchyme-to-epithelium signaling contributes to the development of essentially all organs, glands, and limbs. Structural analysis has shown that members of the FGF7 subfamily achieve their restricted specificity for FGFR1b/FGFR2b by engaging in specific contacts with two alternatively spliced loop regions in the immunoglobulin-like domain 3 (D3) of these receptors. Weak basal receptor-binding affinity further constrains the FGF7 subfamily’s specificity for FGFR1b/2b. In this review, we elaborate on the structural determinants of FGF7 subfamily receptor-binding specificity, and discuss how affinity differences among the four members for the heparin sulfate (HS) co-receptor contribute to their disparate biological activities.
Collapse
Affiliation(s)
- Allen Zinkle
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, United States
| | - Moosa Mohammadi
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, United States
| |
Collapse
|
34
|
Zhao M, Zhuo ML, Zheng X, Su X, Meric-Bernstam F. FGFR1β is a driver isoform of FGFR1 alternative splicing in breast cancer cells. Oncotarget 2019; 10:30-44. [PMID: 30713601 PMCID: PMC6343755 DOI: 10.18632/oncotarget.26530] [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: 10/02/2018] [Accepted: 12/16/2018] [Indexed: 01/11/2023] Open
Abstract
Abnormal FGFR1 alternative splicing is correlated with tumorigenicity and poor prognosis in several tumor types. We sought to determine the roles of FGFR1α and FGFR1β variants in breast cancer. TCGA samples and cell lines were analyzed for FGFR1α/FGFR1β expression. MCF-10A cells were used to overexpress these variants. Cell growth and transformation were assessed by SRB, colony formation, 3D-Matrigel, soft agar, cell motility assays. In TCGA, compared to FGFR1 non-amplified samples, FGFR1-amplified samples had significantly higher FGFR1α but not FGFR1β levels. FGFR1β expression levels and FGFR1β/FGFR1α ratio were higher in basal subtype samples than in ER-positive/luminal samples in both TCGA and breast cancer cell lines. Both FGFR1α and FGFR1β induced transformation of MCF-10A cells. However, only FGFR1β-expressing cells, not FGFR1α, enhanced cell growth and cell motility. Cells with higher FGFR1β levels and FGFR1β/FGFR1α ratio were more sensitive to FGFR inhibitor BGJ-398. Interestingly, in ER-negative cells, FGFR inhibitors decreased FGFR1β levels, likely by increasing expression of splicing repressor PTBP1. In ER-positive cells, estrogen treatment increased FGFR1β levels by decreasing PTBP1 expression, which was blocked by 4-OHT. Lastly, combination treatment with BGJ-398 and 4-OHT synergistically inhibited cell survival. These findings suggest that FGFR1 alternative FGFR1α/FGFR1β splicing plays an important role in breast cancer.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming-Lei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Medical Oncology-I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
35
|
Farrell B, Breeze AL. Structure, activation and dysregulation of fibroblast growth factor receptor kinases: perspectives for clinical targeting. Biochem Soc Trans 2018; 46:1753-1770. [PMID: 30545934 PMCID: PMC6299260 DOI: 10.1042/bst20180004] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 01/22/2023]
Abstract
The receptor tyrosine kinase family of fibroblast growth factor receptors (FGFRs) play crucial roles in embryonic development, metabolism, tissue homeostasis and wound repair via stimulation of intracellular signalling cascades. As a consequence of FGFRs' influence on cell growth, proliferation and differentiation, FGFR signalling is frequently dysregulated in a host of human cancers, variously by means of overexpression, somatic point mutations and gene fusion events. Dysregulation of FGFRs is also the underlying cause of many developmental dysplasias such as hypochondroplasia and achondroplasia. Accordingly, FGFRs are attractive pharmaceutical targets, and multiple clinical trials are in progress for the treatment of various FGFR aberrations. To effectively target dysregulated receptors, a structural and mechanistic understanding of FGFR activation and regulation is required. Here, we review some of the key research findings from the last couple of decades and summarise the strategies being explored for therapeutic intervention.
Collapse
Affiliation(s)
- Brendan Farrell
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Alexander L Breeze
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
36
|
Liu Y, Yu F, Zhang B, Zhou M, Bei Y, Zhang Y, Tang J, Yang Y, Huang Y, Xiang Q, Zhao Y, Liang Q, Liu Y. Improving the protective effects of aFGF for peripheral nerve injury repair using sulfated chitooligosaccharides. Asian J Pharm Sci 2018; 14:511-520. [PMID: 32104478 PMCID: PMC7032102 DOI: 10.1016/j.ajps.2018.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 08/17/2018] [Accepted: 09/27/2018] [Indexed: 01/07/2023] Open
Abstract
Injury to the peripheral nerves can result in temporary or life-long neuronal dysfunction and subsequent economic or social disability. Acidic fibroblast growth factor (aFGF) promotes the growth and survival of neurons and is a possible treatment for peripheral nerve injury. Yet, the actual therapeutic utility of aFGF is limited by its short half-life and instability in vivo. In the present study, we prepared sulfated chitooligosaccharides (SCOS), which have heparin-like properties, to improve the bioactivity of aFGF. We investigated the protective effects of SCOS with or without aFGF on RSC96 cells exposed to Na2S2O4 hypoxia/reoxygenation injury. Cell viability was measured by MTT assay and cytotoxicity induced by Na2S2O4 was assessed by lactate dehydrogenase (LDH) release into the culture medium. Pretreatment with aFGF and SCOS dramatically decreased LDH release after injury compared to pretreatment with aFGF or SCOS alone. We subsequently prepared an aFGF/SCOS thermo-sensitive hydrogel with poloxamer and examined its effects in vivo. Paw withdrawal thresholds and thermal withdrawal latencies were measured in rats with sciatic nerve injury. Local injection of the aFGF/SCOS hydrogels (aFGF: 40, 80 µg/kg) increased the efficiency of sciatic nerve repair compared to aFGF (80 µg/kg) hydrogel alone. Especially aFGF/SCOS thermo-sensitive hydrogel decreased paw withdrawal thresholds from 117.75 ± 8.38 (g, 4 d) to 65.74 ± 3.39 (g, 10 d), but aFGF alone group were 140.58 ± 27.54 (g, 4 d) to 89.12 ± 5.60 (g, 10 d) (aFGF dose was 80 µg/kg, P < 0.05, n = 8). The thermal withdrawal latencies decreased from 11.61 ± 2.26 (s, 4 d) to 2.37 ±0.67 (s, 10 d). However, aFGF alone group were from 17.69 ± 1.47 (s, 4 d) to 4.65 ± 1.73 (s, 10 d) (P < 0.05, n = 8). Furthermore, the aFGF/SCOS hydrogels also exhibited good biocompatibility in mice. In summary, SCOS improved the protective effects of aFGF in RSC96 cells injured with Na2S2O4 and increased the efficiency of nerve repair and recovery of function in rats with sciatic nerve injury. These findings pave an avenue for the development of novel prophylactic and therapeutic strategies for peripheral nerve injury.
Collapse
Affiliation(s)
- Yanmei Liu
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Fenglin Yu
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Beibei Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Meng Zhou
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yu Bei
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yifan Zhang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Jianzhong Tang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Yan Yang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China.,College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China.,College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yueping Zhao
- Department of Stomatology, Jinan University Medical College, Guangzhou 510632, China
| | - Qian Liang
- Department of Stomatology, Jinan University Medical College, Guangzhou 510632, China
| | - Yang Liu
- Department of Stomatology, Jinan University Medical College, Guangzhou 510632, China
| |
Collapse
|
37
|
Collins LE, Troeberg L. Heparan sulfate as a regulator of inflammation and immunity. J Leukoc Biol 2018; 105:81-92. [PMID: 30376187 DOI: 10.1002/jlb.3ru0618-246r] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Heparan sulfate is found on the surface of most cell types, as well as in basement membranes and extracellular matrices. Its strong anionic properties and highly variable structure enable this glycosaminoglycan to provide binding sites for numerous protein ligands, including many soluble mediators of the immune system, and may promote or inhibit their activity. The formation of ligand binding sites on heparan sulfate (HS) occurs in a tissue- and context-specific fashion through the action of several families of enzymes, most of which have multiple isoforms with subtly different specificities. Changes in the expression levels of these biosynthetic enzymes occur in response to inflammatory stimuli, resulting in structurally different HS and acquisition or loss of binding sites for immune mediators. In this review, we discuss the multiple roles for HS in regulating immune responses, and the evidence for inflammation-associated changes to HS structure.
Collapse
Affiliation(s)
- Laura E Collins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Linda Troeberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Stehbens SJ, Ju RJ, Adams MN, Perry SR, Haass NK, Bryant DM, Pollock PM. FGFR2-activating mutations disrupt cell polarity to potentiate migration and invasion in endometrial cancer cell models. J Cell Sci 2018; 131:jcs.213678. [PMID: 30002137 DOI: 10.1242/jcs.213678] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/24/2018] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases that control a diverse range of biological processes during development and in adult tissues. We recently reported that somatic FGFR2 mutations are associated with shorter survival in endometrial cancer. However, little is known about how these FGFR2 mutations contribute to endometrial cancer metastasis. Here, we report that expression of the activating mutations FGFR2N550K and FGFR2Y376C in an endometrial cancer cell model induce Golgi fragmentation, and loss of polarity and directional migration. In mutant FGFR2-expressing cells, this was associated with an inability to polarise intracellular pools of FGFR2 towards the front of migrating cells. Such polarization defects were exacerbated in three-dimensional culture, where FGFR2 mutant cells were unable to form well-organised acini, instead undergoing exogenous ligand-independent invasion. Our findings uncover collective cell polarity and invasion as common targets of disease-associated FGFR2 mutations that lead to poor outcome in endometrial cancer patients.
Collapse
Affiliation(s)
- Samantha J Stehbens
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia .,The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Robert J Ju
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia.,The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Mark N Adams
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Samuel R Perry
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Nikolas K Haass
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - David M Bryant
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Pamela M Pollock
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| |
Collapse
|
39
|
Abstract
Unlike for adenocarcinomas of the lung, no molecular targeted therapies have yet been developed for squamous cell lung cancers, because targetable oncogenic aberrations are scarce in this tumor type. Recent discoveries have established that the fibroblast growth factor (FGF) signaling pathway plays a fundamental role in cancer development by supporting tumor angiogenesis and cancer cell proliferation via different mechanisms. Through comprehensive genomic studies, aberrations in the FGF pathway have been identified in various tumor types, including squamous cell lung cancer, making FGF receptor (FGFR) a potentially druggable target in this malignancy. Several multi-targeted tyrosine kinase inhibitors include FGFR in their target spectrum and a number of these compounds have been approved for clinical use in different cancers. Novel agents selectively targeting FGFRs have been developed and are currently under investigation in clinical trials, showing promising results. This article reviews FGFR aberrations and the clinical data involving selective and multikinase FGFR inhibitors in squamous cell lung cancer.
Collapse
|
40
|
Hui Q, Jin Z, Li X, Liu C, Wang X. FGF Family: From Drug Development to Clinical Application. Int J Mol Sci 2018; 19:ijms19071875. [PMID: 29949887 PMCID: PMC6073187 DOI: 10.3390/ijms19071875] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/17/2018] [Accepted: 06/21/2018] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor (FGF) belongs to a large family of growth factors. FGFs use paracrine or endocrine signaling to mediate a myriad of biological and pathophysiological process, including angiogenesis, wound healing, embryonic development, and metabolism regulation. FGF drugs for the treatment of burn and ulcer wounds are now available. The recent discovery of the crucial roles of the endocrine-acting FGF19 subfamily in maintaining homeostasis of bile acid, glucose, and phosphate further extended the activity profile of this family. Here, the applications of recombinant FGFs for the treatment of wounds, diabetes, hypophosphatemia, the development of FGF receptor inhibitors as anti-neoplastic drugs, and the achievements of basic research and applications of FGFs in China are reviewed.
Collapse
Affiliation(s)
- Qi Hui
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
| | - Zi Jin
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
| | - Xiaokun Li
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
- Key Laboratory Biotechnology Pharmaceutical Engineering, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
| | - Changxiao Liu
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, 308 Anshan West Road, Tianjin 300193, China.
| | - Xiaojie Wang
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
- Key Laboratory Biotechnology Pharmaceutical Engineering, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China.
| |
Collapse
|
41
|
Kappert F, Sreeramulu S, Jonker HRA, Richter C, Rogov VV, Proschak E, Hargittay B, Saxena K, Schwalbe H. Structural Characterization of the Interaction of the Fibroblast Growth Factor Receptor with a Small Molecule Allosteric Inhibitor. Chemistry 2018; 24:7861-7865. [DOI: 10.1002/chem.201801770] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Franziska Kappert
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
| | - Sridhar Sreeramulu
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
| | - Hendrik R. A. Jonker
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
| | - Christian Richter
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
| | - Vladimir V. Rogov
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Biophysical Chemistry; Max von Laue-Straße 9 60438 Frankfurt am Main Germany
| | - Ewgenij Proschak
- Goethe University; Institute of Pharmaceutical Chemistry; Max von Laue-Straße 9 60438 Frankfurt Germany
| | - Bruno Hargittay
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
| | - Krishna Saxena
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
- German Cancer Research Center (DKFZ); Partner facility Frankfurt/Mainz
| | - Harald Schwalbe
- Goethe University; Center for Biomolecular Magnetic Resonance (BMRZ); Institute for Organic Chemistry and Chemical Biology; Max von Laue-Straße 7 60438 Frankfurt am Main Germany
- German Cancer Research Center (DKFZ); Partner facility Frankfurt/Mainz
| |
Collapse
|
42
|
Zhou W, Du X, Song F, Zheng H, Chen K, Zhang W, Yang J. Prognostic roles for fibroblast growth factor receptor family members in malignant peripheral nerve sheath tumor. Oncotarget 2017; 7:22234-44. [PMID: 26993773 PMCID: PMC5008358 DOI: 10.18632/oncotarget.8067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/24/2016] [Indexed: 01/19/2023] Open
Abstract
Background Malignant peripheral nerve sheath tumors (MPNST) are rare, highly malignant, and poorly understood sarcomas. The often poor outcome of MPNST highlights the necessity of identifying prognostic predictors for this aggressive sarcoma. Here, we investigate the role of fibroblast growth factor receptor (FGFR) family members in human MPNSTs. Results aCGH and bioinformatics analysis identified frequent amplification of the FGFR1 gene. FISH analysis revealed that 26.9% MPNST samples had amplification of FGFR1, with both focal and polysomy patterns observed. IHC identified that FGFR1 protein expression was positively correlated with FGFR1 gene amplification. High expression of FGFR1 protein was associated with better overall survival (OS) and was an independent prognostic predictor for OS of MPNST patients. Additionally, combined expression of FGFR1 and FGFR2 protein characterized a subtype of MPNST with better OS. FGFR4 protein was expressed 82.3% of MPNST samples, and was associated with poor disease-free survival. Materials and Methods We performed microarray-based comparative genomic hybridization (aCGH) profiling of two cohorts of primary MPNST tissue samples including 25 patients treated at The University of Texas MD Anderson Cancer Center and 26 patients from Tianjin Medical University Cancer Institute and Hospital. Fluorescence in situ hybridization (FISH) was used to validate the gene amplification detected by aCGH analysis. Another cohort of 63 formalin-fixed paraffin-embedded MPNST samples (including 52 samples for FISH assay) was obtained to explore FGFR1, 2, 3, and 4 protein expression by immunohistochemical (IHC) analysis. Conclusions Our integrated genomic and molecular studies provide evidence that FGFRs play different prognostic roles in MPNST.
Collapse
Affiliation(s)
- Wenya Zhou
- Department of Bone and Soft Tissue Tumor and Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China
| | - Xiaoling Du
- Department of Diagnostics, Tianjin Medical University, Tianjin 300061, People's Republic of China
| | - Fengju Song
- National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China.,Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China
| | - Hong Zheng
- National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China.,Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China
| | - Kexin Chen
- National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China.,Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China
| | - Wei Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030 USA
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor and Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China.,National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, People's Republic of China
| |
Collapse
|
43
|
Lim W, Bae H, Bazer FW, Song G. Stimulatory effects of fibroblast growth factor 2 on proliferation and migration of uterine luminal epithelial cells during early pregnancy. Biol Reprod 2017; 96:185-198. [PMID: 28395342 DOI: 10.1095/biolreprod.116.142331] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/10/2016] [Indexed: 01/11/2023] Open
Abstract
Fibroblast growth factor 2 (FGF2) is a mitogen that induces proliferation, differentiation, and migration of cells, as well as angiogenesis and carcinogenesis via autocrine or paracrine actions. Fibroblast growth factor 2 expression is abundant in porcine conceptuses and endometrium during the estrous cycle and peri-implantation period of pregnancy. However, its intracellular actions in uterine epithelial cells have not been reported. The results of this study indicated abundant expression of FGFR1 and FGFR2 predominantly in uterine luminal and glandular epithelia during early pregnancy and that their expression decreased with increasing parity of the sows. Treatment of porcine uterine luminal epithelial (pLE) cells with FGF2 increased proliferation and DNA replication based on increases in proliferating cell nuclear antigen (PCNA) and initiation of G1/S phase progression. In addition, FGF2 increases phosphorylation of AKT, P70S6K, S6, ERK1/2, JNK, P38, and P90RSK in a time-dependent manner, and increases in their expression was suppressed by Wortmannin (a phosphatidylinositol 3-kinase [PI3K] inhibitor), U0126 (an ERK1/2 inhibitor), SP600125 (a JNK inhibitor), and SB203580 (a P38 inhibitor) based on western blot analyses. Also, the abundance of cytoplasmic p-AKT protein was decreased by Wortmannin and U0126, and p-ERK1/2 protein was reduced only by U0126. Furthermore, inhibition of each signal transduction protein reduced the ability of FGF2 to stimulate proliferation and migration of pLE cells. Collectively, these results indicate that activation of FGFR1 and FGFR2 by uterine- and endometrial-derived FGF2 stimulates PI3K/AKT and mitogen-activated protein kinase pathways for development of the porcine uterus and improvement of litter size.
Collapse
Affiliation(s)
- Whasun Lim
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hyocheol Bae
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Fuller W Bazer
- Department of Animal Science, Center for Animal Biotechnology and Genomics, Texas A&M University, Texas, USA
| | - Gwonhwa Song
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
44
|
Egan JB, Marks DL, Hogenson TL, Vrabel AM, Sigafoos AN, Tolosa EJ, Carr RM, Safgren SL, Enriquez Hesles E, Almada LL, Romecin-Duran PA, Iguchi E, Ala’Aldeen A, Kocher JPA, Oliver GR, Prodduturi N, Mead DW, Hossain A, Huneke NE, Tagtow CM, Ailawadhi S, Ansell SM, Banck MS, Bryce AH, Carballido EM, Chanan-Khan AA, Curtis KK, Resnik E, Gawryletz CD, Go RS, Halfdanarson TR, Ho TH, Joseph RW, Kapoor P, Mansfield AS, Meurice N, Nageswara Rao AA, Nowakowski GS, Pardanani A, Parikh SA, Cheville JC, Feldman AL, Ramanathan RK, Robinson SI, Tibes R, Finnes HD, McCormick JB, McWilliams RR, Jatoi A, Patnaik MM, Silva AC, Wieben ED, McAllister TM, Rumilla KM, Kerr SE, Lazaridis KN, Farrugia G, Stewart AK, Clark KJ, Kennedy EJ, Klee EW, Borad MJ, Fernandez-Zapico ME. Molecular Modeling and Functional Analysis of Exome Sequencing–Derived Variants of Unknown Significance Identify a Novel, Constitutively Active FGFR2 Mutant in Cholangiocarcinoma. JCO Precis Oncol 2017; 2017. [PMID: 30761385 PMCID: PMC6369924 DOI: 10.1200/po.17.00018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Genomic testing has increased the quantity of information available to oncologists. Unfortunately, many identified sequence alterations are variants of unknown significance (VUSs), which thus limit the clinician’s ability to use these findings to inform treatment. We applied a combination of in silico prediction and molecular modeling tools and laboratory techniques to rapidly define actionable VUSs. Materials and Methods Exome sequencing was conducted on 308 tumors from various origins. Most single nucleotide alterations within gene coding regions were VUSs. These VUSs were filtered to identify a subset of therapeutically targetable genes that were predicted with in silico tools to be altered in function by their variant sequence. A subset of receptor tyrosine kinase VUSs was characterized by laboratory comparison of each VUS versus its wild-type counterpart in terms of expression and signaling activity. Results The study identified 4,327 point mutations of which 3,833 were VUSs. Filtering for mutations in genes that were therapeutically targetable and predicted to affect protein function reduced these to 522 VUSs of interest, including a large number of kinases. Ten receptor tyrosine kinase VUSs were selected to explore in the laboratory. Of these, seven were found to be functionally altered. Three VUSs (FGFR2 F276C, FGFR4 R78H, and KDR G539R) showed increased basal or ligand-stimulated ERK phosphorylation compared with their wild-type counterparts, which suggests that they support transformation. Treatment of a patient who carried FGFR2 F276C with an FGFR inhibitor resulted in significant and sustained tumor response with clinical benefit. Conclusion The findings demonstrate the feasibility of rapid identification of the biologic relevance of somatic mutations, which thus advances clinicians’ ability to make informed treatment decisions.
Collapse
Affiliation(s)
- Jan B. Egan
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - David L. Marks
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Tara L. Hogenson
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Anne M. Vrabel
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Ashley N. Sigafoos
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Ezequiel J. Tolosa
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Ryan M. Carr
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Stephanie L. Safgren
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Elisa Enriquez Hesles
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Luciana L. Almada
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Paola A. Romecin-Duran
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Eriko Iguchi
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Aryan Ala’Aldeen
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Jean-Pierre A. Kocher
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Gavin R. Oliver
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Naresh Prodduturi
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - David W. Mead
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Asif Hossain
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Norine E. Huneke
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Colleen M. Tagtow
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Sikander Ailawadhi
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Stephen M. Ansell
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Michaela S. Banck
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Alan H. Bryce
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Estrella M. Carballido
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Asher A. Chanan-Khan
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Kelly K. Curtis
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Ernesto Resnik
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Chelsea D. Gawryletz
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Ronald S. Go
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Thorvardur R. Halfdanarson
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Thai H. Ho
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Richard W. Joseph
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Prashant Kapoor
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Aaron S. Mansfield
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Nathalie Meurice
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Amulya A. Nageswara Rao
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Grzegorz S. Nowakowski
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Animesh Pardanani
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Sameer A. Parikh
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - John C. Cheville
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Andrew L. Feldman
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Ramesh K. Ramanathan
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Steven I. Robinson
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Raoul Tibes
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Heidi D. Finnes
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Jennifer B. McCormick
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Robert R. McWilliams
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Aminah Jatoi
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Mrinal M. Patnaik
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Alvin C. Silva
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Eric D. Wieben
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | | | | | | | | | - Gianrico Farrugia
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - A. Keith Stewart
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Karl J. Clark
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Eileen J. Kennedy
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Eric W. Klee
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Mitesh J. Borad
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| | - Martin E. Fernandez-Zapico
- David L. Marks, Tara L. Hogenson, Anne M. Vrabel, Ashley N. Sigafoos, Ezequiel J. Tolosa, Ryan M. Carr, Stephanie L. Safgren, Elisa Enriquez Hesles, Luciana L. Almada, Paola A. Romecin-Duran, Eriko Iguchi, Aryan Ala’Aldeen, Jean-Pierre A. Kocher, Gavin R. Oliver, Naresh Prodduturi, David W. Mead, Asif Hossain, Norine E. Huneke, Colleen M. Tagtow, Sikander Ailawadhi, Stephen M. Ansell, Michaela S. Banck, Asher A. Chanan-Khan, Ronald S. Go, Thorvardur R. Halfdanarson, Richard W. Joseph, Prashant Kapoor,
| |
Collapse
|
45
|
Opalinski L, Szczepara M, Sokolowska-Wedzina A, Zakrzewska M, Otlewski J. The autoinhibitory function of D1 domain of FGFR1 goes beyond the inhibition of ligand binding. Int J Biochem Cell Biol 2017; 89:193-198. [PMID: 28652212 DOI: 10.1016/j.biocel.2017.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 01/12/2023]
Abstract
Fibroblast growth factors (FGFs) and their plasma membrane-localized receptors (FGFRs) transduce signals that regulate developmental processes and metabolism. In numerous cancer types genetic aberrations of FGFR1 lead to its uncontrolled activation. To circumvent the unrestrained signal transduction, several intramolecular inhibitory mechanisms within FGFR1 have evolved. In vitro experiments with receptor truncation have demonstrated that the N-terminal D1 domain of FGFR1 negatively regulates ligand binding to the receptor. Here, we show that D1-specific monovalent antibody fragments can activate FGFR1 and its downstream signaling cascades in the absence of ligand. These data suggest that the D1 domain of FGFR1 may play autoinhibitory role not only by controlling ligand binding, but also by regulating the overall conformation of FGFR1, keeping it in a state that disfavors autoactivation in the absence of its cognate growth factor.
Collapse
Affiliation(s)
- Lukasz Opalinski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Martyna Szczepara
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Aleksandra Sokolowska-Wedzina
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
46
|
Wang S, Ding Z. Fibroblast growth factor receptors in breast cancer. Tumour Biol 2017; 39:1010428317698370. [PMID: 28459213 DOI: 10.1177/1010428317698370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Fibroblast growth factor receptors are growth factor receptor tyrosine kinases, exerting their roles in embryogenesis, tissue homeostasis, and development of breast cancer. Recent genetic studies have identified some subtypes of fibroblast growth factor receptors as strong genetic loci associated with breast cancer. In this article, we review the recent epidemiological findings and experiment results of fibroblast growth factor receptors in breast cancer. First, we summarized the structure and physiological function of fibroblast growth factor receptors in humans. Then, we discussed the common genetic variations in fibroblast growth factor receptors that affect breast cancer risk. In addition, we also introduced the potential roles of each fibroblast growth factor receptors isoform in breast cancer. Finally, we explored the potential therapeutics targeting fibroblast growth factor receptors for breast cancer. Based on the biological mechanisms of fibroblast growth factor receptors leading to the pathogenesis in breast cancer, targeting fibroblast growth factor receptors may provide new opportunities for breast cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shuwei Wang
- Department of General Surgery, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Zhongyang Ding
- Department of General Surgery, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, P.R. China
| |
Collapse
|
47
|
Cheng W, Wang M, Tian X, Zhang X. An overview of the binding models of FGFR tyrosine kinases in complex with small molecule inhibitors. Eur J Med Chem 2016; 126:476-490. [PMID: 27914362 DOI: 10.1016/j.ejmech.2016.11.052] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/19/2016] [Accepted: 11/07/2016] [Indexed: 11/26/2022]
Abstract
The fibroblast growth factor receptor (FGFR) family receptor tyrosine kinase (RTK) includes four structurally related members, termed as FGFR1, FGFR2, FGFR3, and FGFR4. Given its intimate role in the progression of several solid tumors, excessive FGFR signaling provides an opportunity for anticancer therapy. Along with extensive pharmacological studies validating the therapeutic potential of targeting the FGFRs for cancer treatment, co-crystal structures of FGFRs/inhibitors are continuously coming up to study the mechanism of actions and explore new inhibitors. Herein, we review the reported co-crystals of FGFRs in complex with the corresponding inhibitors, main focusing our attention on the binding models and the pharmacological activities of the inhibitors.
Collapse
Affiliation(s)
- Weiyan Cheng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mixiang Wang
- Department of Pharmacy, The First Affiliated Hospital of Nanyang Medical College, Nanyang 473000, China
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
48
|
Zhou WY, Zheng H, Du XL, Yang JL. Characterization of FGFR signaling pathway as therapeutic targets for sarcoma patients. Cancer Biol Med 2016; 13:260-8. [PMID: 27458533 PMCID: PMC4944539 DOI: 10.20892/j.issn.2095-3941.2015.0102] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The fibroblast growth factor receptor (FGFR) family plays important roles in regulating cell growth, proliferation, survival, differentiation and angiogenesis. Deregulation of the FGF/FGFR signaling pathway has been associated with multiple development syndromes and cancers, and thus therapeutic strategies targeting FGFs and FGFR in human cancer are currently being explored. However, few studies on the FGF/FGFR pathway have been conducted in sarcoma, which has a poor outcome with traditional treatments such as surgery, chemotherapy, and radiotherapy. Hence, in the present review, we provide an overview of the role of the FGF/FGFR pathway signal in sarcoma and FGFR inhibitors, which might be new targets for the treatment of sarcomas according to recent research.
Collapse
Affiliation(s)
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiao-Ling Du
- Department of Diagnostics, Tianjin Medical University, Tianjin 300061, China
| | | |
Collapse
|
49
|
Hallinan N, Finn S, Cuffe S, Rafee S, O’Byrne K, Gately K. Targeting the fibroblast growth factor receptor family in cancer. Cancer Treat Rev 2016; 46:51-62. [DOI: 10.1016/j.ctrv.2016.03.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 02/08/2023]
|
50
|
Yeon JH, Heinkel F, Sung M, Na D, Gsponer J. Systems-wide Identification of cis-Regulatory Elements in Proteins. Cell Syst 2016; 2:89-100. [PMID: 27135163 DOI: 10.1016/j.cels.2016.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/15/2015] [Accepted: 02/01/2016] [Indexed: 10/22/2022]
Abstract
Protein interactions in cis that can activate or autoinhibit protein function play an important role in the fine-tuning of regulatory and signaling processes in the cell, but thus far cis-regulatory elements (CREs) in proteins have not been systematically identified and studied. Here, we introduce a computational tool that identifies intrinsically disordered protein segments that contribute to protein function regulation via interactions in cis. We apply this tool to estimate the prevalence of CREs in the human proteome and reveal that cis regulation is enriched in several signaling pathways, including the MAP kinase pathway, for which we provide a detailed map of its "cis regulome." We also show that disease-causing mutations are highly enriched in CREs, but not in motifs that classically mediate protein-protein interactions of disordered protein segments. Our approach should facilitate the discovery and characterization of CREs in proteins and the identification of disease-causing mutations that disrupt protein regulation in cis.
Collapse
Affiliation(s)
- Ju Hun Yeon
- Department of Integrative Bioscience, University of Brain Education, 284-31 Gyocheonjisan-gil, Mokcheon-eup, Dongnam-gu, Cheonan, Chungcheongnam-do 31228, Republic of Korea
| | - Florian Heinkel
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, 2125 East Mall, Vancouver, BC V6T 1Z4, Canada
| | - Minhui Sung
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Dokyun Na
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea; Centre for High-Throughput Biology, University of British Columbia, 2125 East Mall, Vancouver, BC V6T 1Z4, Canada.
| | - Jörg Gsponer
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, 2125 East Mall, Vancouver, BC V6T 1Z4, Canada; Centre for High-Throughput Biology, University of British Columbia, 2125 East Mall, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|