1
|
Zlinska V, Feketova Z, Czyrek A, Chudzian J, Zivkovic ML, Ursachi VC, Dudeja P, Fafilek B, Rynes J, Rico-Llanos G, Koudelka A, Roy T, Biadun M, Raskova V, Svozilova K, Stroblova M, Krzyscik M, Hristova K, Krowarsch D, Foldynova-Trantirkova S, Zakrzewska M, Trantirek L, Krejci P. Specific inhibition of fibroblast growth factor receptor 1 signaling by a DNA aptamer. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102405. [PMID: 39759879 PMCID: PMC11700292 DOI: 10.1016/j.omtn.2024.102405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025]
Abstract
Impaired fibroblast growth factor receptor (FGFR) signaling is associated with many human conditions, including growth disorders, degenerative diseases, and cancer. Current FGFR therapeutics are based on chemical inhibitors of FGFR tyrosine kinase activity (TKIs). However, FGFR TKIs are limited in their target specificity as they generally inhibit all FGFRs and other receptor tyrosine kinases. In the search for specific inhibitors of human FGFR1, we identified VZ23, a DNA aptamer that binds to FGFR1b and FGFR1c with a KD of 55 nM and 162 nM, respectively, but not to the other FGFR variants (FGFR2b, FGFR2c, FGFR3b, FGFR3c, FGFR4). In cells, VZ23 inhibited the activation of downstream FGFR1 signaling and FGFR1-mediated regulation of cellular senescence, proliferation, and extracellular matrix homeostasis. Consistent with the specificity toward FGFR1 observed in vitro, VZ23 did not inhibit FGFR2-4 signaling in cells. We show that the VZ23 inhibits FGFR1 signaling in the presence of cognate fibroblast growth factor (FGF) ligands and its inhibitory activity is linked to its capacity to form unusual G-quadruplex structure. Our data suggest that targeting FGFR1 with DNA aptamers could be an effective alternative to TKIs for treating impaired FGFR1 signaling in human craniosynostoses.
Collapse
Affiliation(s)
- Vladimira Zlinska
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czechia
- National Centre for Biomolecular Research, Masaryk University, 625 00 Brno, Czechia
| | - Zuzana Feketova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
| | - Aleksandra Czyrek
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
| | - Julia Chudzian
- Department of Protein Engineering, University of Wroclaw, 50-383 Wroclaw, Poland
| | | | - Vlad-Constantin Ursachi
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
| | - Pooja Dudeja
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
| | - Bohumil Fafilek
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czechia
| | - Jan Rynes
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czechia
| | - Gustavo Rico-Llanos
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
| | - Adolf Koudelka
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
| | - Tanaya Roy
- Department of Materials Science and Engineering, Institute for NanoBioTechnology and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Martyna Biadun
- Department of Protein Engineering, University of Wroclaw, 50-383 Wroclaw, Poland
| | - Vendula Raskova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
| | - Katerina Svozilova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czechia
| | - Michaela Stroblova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
| | - Mateusz Krzyscik
- Department of Materials Science and Engineering, Institute for NanoBioTechnology and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Daniel Krowarsch
- Department of Protein Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland
| | | | | | - Lukas Trantirek
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czechia
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czechia
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czechia
| |
Collapse
|
2
|
Chen G, Chen L, Li X, Mohammadi M. FGF-based drug discovery: advances and challenges. Nat Rev Drug Discov 2025:10.1038/s41573-024-01125-w. [PMID: 39875570 DOI: 10.1038/s41573-024-01125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
3
|
Allsopp RJ, Klauda JB. Understanding Folding of bFGF and Potential Cellular Protective Mechanisms of Neural Cells. Biochemistry 2025; 64:509-524. [PMID: 39749909 DOI: 10.1021/acs.biochem.4c00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Traumatic brain injury (TBI) is a serious health condition that affects an increasing number of people, especially veterans and athletes. TBI causes serious consequences because of its long-lasting impact on the brain and its alarming frequency of occurrence. Although the brain has some natural protective mechanisms, the processes that trigger them are poorly understood. Fibroblast growth factor (FGF) proteins interact with receptor proteins to protect cells. Gaps in the literature include how basic-FGF (bFGF) is activated by heparin, can heparin-like molecules induce neural protection, and the effect of allosteric binding on bFGF activity. To fill the gap in our understanding, we applied temperature replica exchange to study the influence of heparin binding to bFGF and how mutations in bFGF influence stability. A new favorable binding site was identified by comparing free energies computed from the potential of mean force (PMF). Although the varied sugars studied resulted in different interactions with bFGF compared to heparin, they each produced structural effects similar to those of bFGF that likely facilitate receptor binding and signaling. Our results also demonstrate how point mutations can trigger the same conformational change that is believed to promote favorable interactions with the receptor. A deeper atomic-level understanding of how chemicals are released during TBI is needed to improve the development of new treatments for TBI and could contribute to a better understanding of other diseases.
Collapse
Affiliation(s)
- Robert J Allsopp
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
- Institute for Physical Science and Technology, Biophysics Program, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
4
|
McCaig CD. Electrical Forces Improve Memory in Old Age. Rev Physiol Biochem Pharmacol 2025; 187:453-520. [PMID: 39838022 DOI: 10.1007/978-3-031-68827-0_21] [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] [Indexed: 01/23/2025]
Abstract
This penultimate chapter is based on a single paper published in Nature in 2022. I have used it specifically as an exemplar, in this case to show that memory improvement in old age may be regulated by a multiplicity of electrical forces. However, I include it because I believe that one could pick almost any other substantial single paper and show that a completely disparate set of biological mechanisms similarly depend crucially on multiple electrical forces.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
5
|
de La Bourdonnaye G, Marek M, Ghazalova T, Damborsky J, Pachl P, Brynda J, Stepankova V, Chaloupkova R. Structural analysis of the stable form of fibroblast growth factor 2 - FGF2-STAB. J Struct Biol X 2024; 10:100112. [PMID: 39512606 PMCID: PMC11541812 DOI: 10.1016/j.yjsbx.2024.100112] [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: 07/04/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
Fibroblast growth factor 2 (FGF2) is a signaling protein that plays a significant role in tissue development and repair. FGF2 binds to fibroblast growth factor receptors (FGFRs) alongside its co-factor heparin, which protects FGF2 from degradation. The binding between FGF2 and FGFRs induces intracellular signaling pathways such as RAS-MAPK, PI3K-AKT, and STAT. FGF2 has strong potential for application in cell culturing, wound healing, and cosmetics but the potential is severely limited by its low protein stability. The thermostable variant FGF2-STAB was constructed by computer-assisted protein engineering to overcome the natural limitation of FGF2. Previously reported characterization of FGF2-STAB revealed an enhanced ability to induce MAP/ERK signaling while having a lower dependence on heparin when compared with FGF2-wt. Here we report the crystal structure of FGF2-STAB solved at 1.3 Å resolution. Protein stabilization is achieved by newly formed hydrophobic interactions, polar contacts, and one additional hydrogen bond. The overall structure of FGF2-STAB is similar to FGF2-wt and does not reveal information on the experimentally observed lower dependence on heparin. A noticeable difference in flexibility in the receptor binding region can explain the differences in signaling between FGF2-STAB and its wild-type counterpart. Our structural analysis provided molecular insights into the stabilization and unique biological properties of FGF2-STAB.
Collapse
Affiliation(s)
- Gabin de La Bourdonnaye
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Enantis Ltd., Biotechnology Incubator INBIT, Brno, Czech Republic
| | - Martin Marek
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Tereza Ghazalova
- Enantis Ltd., Biotechnology Incubator INBIT, Brno, Czech Republic
| | - Jiri Damborsky
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Petr Pachl
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jiri Brynda
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Radka Chaloupkova
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Enantis Ltd., Biotechnology Incubator INBIT, Brno, Czech Republic
| |
Collapse
|
6
|
Hoshiyama J, Hayata Y, Eguchi A, Morimoto J, Ueki R, Sando S. Analysis of cell signaling profiles induced by DNA aptamer-based FGFR1 agonist. ANAL SCI 2024; 40:2251-2258. [PMID: 39249203 PMCID: PMC11588945 DOI: 10.1007/s44211-024-00660-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
DNA aptamers have attracted attention as an alternative modality for biomolecules due to their excellent target binding specificity and thermal stability, and they are also expected to be applied as artificial agonists for receptor proteins. DNA aptamer agonist TD0 targeting the receptor of fibroblast growth factor (FGFR), which plays an important role in the fields of wound healing and regenerative medicine, has been reported to induce cellular responses as well as its native ligands. However, it was also noted that there were some different responses upon long-term stimulation, suggesting that the intracellular signals induced by DNA aptamer agonist TD0 are different from those of natural ligands. In this paper, we comprehensively analyzed the intracellular signals induced by DNA aptamer agonist TD0 targeting FGFR1, and compared them with those by natural protein ligand FGF2. It was found that the intracellular signals were highly similar for short-term stimulation. On the other hand, the receptor and the downstream cellular signals showed different activation behaviors for long-time stimulation. Evaluating the stability and sustained activity of DNA aptamer agonist TD0 and FGF2 in the medium suggested that ligand stability may be important in properly regulating cellular responses.
Collapse
Affiliation(s)
- Junya Hoshiyama
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuri Hayata
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Akihiro Eguchi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Jumpei Morimoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ryosuke Ueki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
7
|
Wiens KR, Wasti N, Ulloa OO, Klegeris A. Diversity of Microglia-Derived Molecules with Neurotrophic Properties That Support Neurons in the Central Nervous System and Other Tissues. Molecules 2024; 29:5525. [PMID: 39683685 DOI: 10.3390/molecules29235525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Microglia, the brain immune cells, support neurons by producing several established neurotrophic molecules including glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Modern analytical techniques have identified numerous phenotypic states of microglia, each associated with the secretion of a diverse set of substances, which likely include not only canonical neurotrophic factors but also other less-studied molecules that can interact with neurons and provide trophic support. In this review, we consider the following eight such candidate cytokines: oncostatin M (OSM), leukemia inhibitory factor (LIF), activin A, colony-stimulating factor (CSF)-1, interleukin (IL)-34, growth/differentiation factor (GDF)-15, fibroblast growth factor (FGF)-2, and insulin-like growth factor (IGF)-2. The available literature provides sufficient evidence demonstrating murine cells produce these cytokines and that they exhibit neurotrophic activity in at least one neuronal model. Several distinct types of neurotrophic activity are identified that only partially overlap among the cytokines considered, reflecting either their distinct intrinsic properties or lack of comprehensive studies covering the full spectrum of neurotrophic effects. The scarcity of human-specific studies is another significant knowledge gap revealed by this review. Further studies on these potential microglia-derived neurotrophic factors are warranted since they may be used as targeted treatments for diverse neurological disorders.
Collapse
Affiliation(s)
- Kennedy R Wiens
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Naved Wasti
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Omar Orlando Ulloa
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
8
|
Qi Y, Deng SM, Wang KS. Receptor tyrosine kinases in breast cancer treatment: unraveling the potential. Am J Cancer Res 2024; 14:4172-4196. [PMID: 39417188 PMCID: PMC11477839 DOI: 10.62347/kivs3169] [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: 02/17/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Breast cancer is a multifactorial disease driven by acquired genetic and epigenetic changes that lead to aberrant regulation of cellular signaling pathways. Receptor tyrosine kinases (RTKs), a class of critical receptors, are involved in the initiation and progression of breast cancer. RTKs are cell surface receptors with unique structures and biological characteristics, which respond to environmental signals by initiating signaling cascades such as the mitogen-activated protein kinase (MAPK) pathway, Janus kinase (JAK)/signal transducer, activator of transcription (STAT) pathway, and phosphoinositide 3-kinase (PI3K)/AKT pathway. The critical role of RTKs makes them suitable targets for breast cancer treatment. Targeted therapies against RTKs have been developed in recent years, evaluated in clinical trials, and approved for several cancer types, including breast cancer. However, breast cancer displays molecular heterogeneity and exhibits different therapeutic responses to various drug types, leading to limited effectiveness of targeted therapy against RTKs. In this review, we summarize the structural and functional characteristics of selected RTKs and discuss the mechanisms and current status of drug therapy involving different protein tyrosine kinases in breast cancer progression.
Collapse
Affiliation(s)
- Yu Qi
- Department of Pathology, School of Basic Medical Sciences, Central South UniversityChangsha, Hunan, China
| | - Shu-Min Deng
- Department of Pathology, School of Basic Medical Sciences, Central South UniversityChangsha, Hunan, China
| | - Kuan-Song Wang
- Department of Pathology, School of Basic Medical Sciences, Central South UniversityChangsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| |
Collapse
|
9
|
Tonni E, Oltrecolli M, Pirola M, Tchawa C, Roccabruna S, D'Agostino E, Matranga R, Piombino C, Pipitone S, Baldessari C, Bacchelli F, Dominici M, Sabbatini R, Vitale MG. New Advances in Metastatic Urothelial Cancer: A Narrative Review on Recent Developments and Future Perspectives. Int J Mol Sci 2024; 25:9696. [PMID: 39273642 PMCID: PMC11395814 DOI: 10.3390/ijms25179696] [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: 08/07/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
The standard of care for advanced or metastatic urothelial carcinoma (mUC) was historically identified with platinum-based chemotherapy. Thanks to the advances in biological and genetic knowledge and technologies, new therapeutic agents have emerged in this setting recently: the immune checkpoint inhibitors and the fibroblast growth factor receptor inhibitors as the target therapy for patients harboring alterations in the fibroblast growth factor receptor (FGFR) pathway. However, chasing a tumor's tendency to recur and progress, a new class of agents has more recently entered the scene, with promising results. Antibody-drug conjugates (ADCs) are in fact the latest addition, with enfortumab vedotin being the first to receive accelerated approval by the U.S. Food and Drug Administration in December 2019, followed by sacituzumab govitecan. Many other ADCs are still under investigation. ADCs undoubtedly represent the new frontier, with the potential of transforming the management of mUC treatment in the future. Therefore, we reviewed the landscape of mUC treatment options, giving an insight into the molecular basis and mechanisms, and evaluating new therapeutic strategies in the perspective of more and more personalized treatments.
Collapse
Affiliation(s)
- Elena Tonni
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Marco Oltrecolli
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Marta Pirola
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Cyrielle Tchawa
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Sara Roccabruna
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Elisa D'Agostino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Rossana Matranga
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Claudia Piombino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Stefania Pipitone
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Cinzia Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Francesca Bacchelli
- Clinical Trials Office, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Roberto Sabbatini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| |
Collapse
|
10
|
An YJ, Jung YE, Lee KW, Kaushal P, Ko IY, Shin SM, Ji S, Yu W, Lee C, Lee WK, Cha K, Lee JH, Cha SS, Yim HS. Structural and biochemical investigation into stable FGF2 mutants with novel mutation sites and hydrophobic replacements for surface-exposed cysteines. PLoS One 2024; 19:e0307499. [PMID: 39236042 PMCID: PMC11376533 DOI: 10.1371/journal.pone.0307499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/06/2024] [Indexed: 09/07/2024] Open
Abstract
Fibroblast growth factor 2 (FGF2) is an attractive biomaterial for pharmaceuticals and functional cosmetics. To improve the thermo-stability of FGF2, we designed two mutants harboring four-point mutations: FGF2-M1 (D28E/C78L/C96I/S137P) and FGF2-M2 (D28E/C78I/C96I/S137P) through bioinformatics, molecular thermodynamics, and molecular modeling. The D28E mutation reduced fragmentation of the FGF2 wild type during preparation, and the substitution of a whale-specific amino acid, S137P, enhanced the thermal stability of FGF2. Surface-exposed cysteines that participate in oligomerization through intermolecular disulfide bond formation were substituted with hydrophobic residues (C78L/C78I and C96I) using the in silico method. High-resolution crystal structures revealed at the atomic level that the introduction of mutations stabilizes each local region by forming more favorable interactions with neighboring residues. In particular, P137 forms CH-π interactions with the side chain indole ring of W123, which seems to stabilize a β-hairpin structure, containing a heparin-binding site of FGF2. Compared to the wild type, both FGF2-M1 and FGF2-M2 maintained greater solubility after a week at 45 °C, with their Tm values rising by ~ 5 °C. Furthermore, the duration for FGF2-M1 and FGF2-M2 to reach 50% residual activity at 45 °C extended to 8.8- and 8.2-fold longer, respectively, than that of the wild type. Interestingly, the hydrophobic substitution of surface-exposed cysteine in both FGF2 mutants makes them more resistant to proteolytic cleavage by trypsin, subtilisin, proteinase K, and actinase than the wild type and the Cys → Ser substitution. The hydrophobic replacements can influence protease resistance as well as oligomerization and thermal stability. It is notable that hydrophobic substitutions of surface-exposed cysteines, as well as D28E and S137P of the FGF2 mutants, were designed through various approaches with structural implications. Therefore, the engineering strategies and structural insights adopted in this study could be applied to improve the stability of other proteins.
Collapse
Affiliation(s)
- Young Jun An
- Marine Biotechnology & Bioresource Research Department, Korea Institute of Ocean Science and Technology, Busan, Republic of Korea
| | - Ye-Eun Jung
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Republic of Korea
| | - Kyeong Won Lee
- Marine Biotechnology & Bioresource Research Department, Korea Institute of Ocean Science and Technology, Busan, Republic of Korea
| | - Prashant Kaushal
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic Korea
| | - In Young Ko
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongiu, Republic of Korea
| | - Seung Min Shin
- Marine Biotechnology & Bioresource Research Department, Korea Institute of Ocean Science and Technology, Busan, Republic of Korea
| | - Sangho Ji
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Wookyung Yu
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Cheolju Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic Korea
| | - Won-Kyu Lee
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongiu, Republic of Korea
| | - Kiweon Cha
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongiu, Republic of Korea
| | - Jung-Hyun Lee
- Marine Biotechnology & Bioresource Research Department, Korea Institute of Ocean Science and Technology, Busan, Republic of Korea
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Republic of Korea
| | - Hyung-Soon Yim
- Marine Biotechnology & Bioresource Research Department, Korea Institute of Ocean Science and Technology, Busan, Republic of Korea
| |
Collapse
|
11
|
Nickle A, Ko S, Merrill AE. Fibroblast growth factor 2. Differentiation 2024; 139:100733. [PMID: 37858405 PMCID: PMC11009566 DOI: 10.1016/j.diff.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/20/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
Fibroblast Growth Factor 2 (FGF2), also known as basic fibroblast growth factor, is a potent stimulator of growth and differentiation in multiple tissues. Its discovery traces back over 50 years ago when it was first isolated from bovine pituitary extracts due to its ability to stimulate fibroblast proliferation. Subsequent studies investigating the genomic structure of FGF2 identified multiple protein isoforms, categorized as the low molecular weight and high molecular weight FGF2. These isoforms arise from alternative translation initiation events and exhibit unique molecular and cellular functions. In this concise review, we aim to provide an overview of what is currently known about the structure, expression, and functions of the FGF2 isoforms within the contexts of development, homeostasis, and disease.
Collapse
Affiliation(s)
- Audrey Nickle
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sebastian Ko
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
12
|
Fish JL. Fibroblast growth factors-An introduction to our primer series. Differentiation 2024; 139:100804. [PMID: 39117549 DOI: 10.1016/j.diff.2024.100804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Affiliation(s)
- Jennifer L Fish
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, USA.
| |
Collapse
|
13
|
Fujii I, Kinoshita R, Akiyama H, Nakamura A, Iwamori K, Fukada SI, Honda H, Shimizu K. Discovery of fibroblast growth factor 2-derived peptides for enhancing mice skeletal muscle satellite cell proliferation. Biotechnol J 2024; 19:e2400278. [PMID: 39212202 DOI: 10.1002/biot.202400278] [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: 04/24/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Skeletal muscle satellite cells (SCs) are essential for muscle regeneration. Their proliferation and differentiation are influenced by fibroblast growth factor (FGF)-2. In this study, we screened for FGF-2-derived peptides that promote SC proliferation. Utilizing photocleavable peptide array technology, a library of 7-residue peptides was synthesized, and its effect on SC proliferation was examined using a mixture of five peptides. The results showed that peptides 1-5 (136%), 21-25 (136%), 26-30 (141%), 31-35 (159%), 71-75 (135%), 76-80 (144%), and 126-130 (137%) significantly increased SC proliferation. Further experiments revealed that peptide 33, CKNGGFF, enhanced SC proliferation. Furthermore, its extended form, peptide 33-13, CKNGGFFLRIHPD, promoted SC proliferation and increased the percentage of Pax7-positive cells, indicating that SCs were maintained in an undifferentiated state. The addition of FGF-2 and peptide 33-13 further induced cell proliferation but did not increase the percentage of Pax7-positive cells. A proliferation assay using an FGF receptor (FGFR) inhibitor suggested that peptide 33-13 acts through the FGFR-mediated and other pathways. Although further research is necessary to explore the mechanisms of action of these peptides and their potential for in vivo and in vitro use, the high sequence conservation of peptides 33 and 33-13 in FGF-2 across multiple species suggests their broad application prospects in biomedical engineering and biotechnology.
Collapse
Affiliation(s)
- Itsuki Fujii
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Remi Kinoshita
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Hirokazu Akiyama
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Ayasa Nakamura
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kanako Iwamori
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| |
Collapse
|
14
|
Kuroyanagi G, Hioki T, Matsushima-Nishiwaki R, Kozawa O, Tokuda H. Gallein increases the fibroblast growth factor 2-elicited osteoprotegerin synthesis in osteoblasts. Biochim Biophys Acta Gen Subj 2024; 1868:130635. [PMID: 38788984 DOI: 10.1016/j.bbagen.2024.130635] [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: 12/03/2023] [Revised: 04/28/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Gallein is known as an inhibitor of Gβγ subunits, but roles of gallein in bone metabolism have not been reported. Fibroblast growth factor 2 (FGF-2) increases angiogenesis and promotes bone regeneration during the early stages of fracture healing. Osteoprotegerin (OPG) secreted by osteoblasts, binds to the receptor activator of nuclear factor-κB (RANK) ligand (RANKL) as a decoy receptor and prevents RANKL from binding to RANK, resulting in the suppression of bone resorption. Our previous report demonstrated that FGF-2 activates the phosphorylation of p38 mitogen-activated protein kinase (MAPK), stress-activated protein kinase/c-Jun N-terminal kinase (JNK), and p44/p42 MAPK in osteoblast-like MC3T3-E1 cells. Additionally, FGF-2-activated phosphorylation of p38 MAPK and JNK but not p44/p42 MAPK is positively involved in OPG synthesis in these cells. This work aimed to investigate the effects of gallein on the FGF-2-elicited OPG synthesis in osteoblast-like MC3T3-E1 cells and the mechanism. Our findings demonstrated that gallein significantly increased the FGF-2-elicited OPG synthesis in MC3T3-E1 cells. By contrast, fluorescein, gallein-like compound that does not bind Gβγ, did not affect the FGF-2-elicited OPG synthesis. Gallein significantly enhanced the FGF-2-induced OPG mRNA expression levels. Gallein did not affect the FGF-2-activated phosphorylation of p38 MAPK and p44/p42 MAPK, but significantly increased the FGF-2-activated phosphorylation of JNK, while fluorescein did not affect JNK phosphorylation. SP600125, a specific JNK inhibitor, strongly inhibited gallein-induced enhancement of FGF-2-induced OPG synthesis and mRNA expression levels. Our results indicated that gallein increases the FGF-2-induced OPG synthesis due to the JNK activation in the osteoblast.
Collapse
Affiliation(s)
- Gen Kuroyanagi
- Department of Orthopaedic Surgery, Nagoya City University, Nagoya 467-8601, Japan; Department of Rehabilitation Medicine, Nagoya City University, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.
| | - Tomoyuki Hioki
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan; Department of Dermatology, Kizawa Memorial Hospital, Minokamo, Gifu 505-0034, Japan
| | - Rie Matsushima-Nishiwaki
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University, Gifu 501-1194, Japan; Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan; Department of Clinical Laboratory, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| |
Collapse
|
15
|
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
|
16
|
Shivgan AT, Marzinek JK, Krah A, Matsudaira P, Verma CS, Bond PJ. Coarse-Grained Model of Glycosaminoglycans for Biomolecular Simulations. J Chem Theory Comput 2024; 20:3308-3321. [PMID: 38358378 DOI: 10.1021/acs.jctc.3c01088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Proteoglycans contain glycosaminoglycans (GAGs) which are negatively charged linear polymers made of repeating disaccharide units of uronic acid and hexosamine units. They play vital roles in numerous physiological and pathological processes, particularly in governing cellular communication and attachment. Depending on their sulfonation state, acetylation, and glycosidic linkages, GAGs belong to different families. The high molecular weight, heterogeneity, and flexibility of GAGs hamper their characterization at atomic resolution, but this may be circumvented via coarse-grained (CG) approaches. In this work, we report a CG model for a library of common GAG types in their isolated or proteoglycan-linked states compatible with version 2.2 (v2.2) of the widely popular CG Martini force field. The model reproduces conformational and thermodynamic properties for a wide variety of GAGs, as well as matching structural and binding data for selected proteoglycan test systems. The parameters developed here may thus be employed to study a range of GAG-containing biomolecular systems, thereby benefiting from the efficiency and broad applicability of the Martini framework.
Collapse
Affiliation(s)
- Aishwary T Shivgan
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543, Singapore
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Jan K Marzinek
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Alexander Krah
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Paul Matsudaira
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543, Singapore
| | - Chandra S Verma
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543, Singapore
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
- School of Biological sciences, Nanyang Technological University, 50 Nanyang Drive, Singapore 637551, Singapore
| | - Peter J Bond
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543, Singapore
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| |
Collapse
|
17
|
Hashimoto U, Fujitani N, Uehara Y, Okamoto H, Saitou A, Ito F, Ariki S, Shiratsuchi A, Hasegawa Y, Takahashi M. N-glycan on N262 of FGFR3 regulates the intracellular localization and phosphorylation of the receptor. Biochim Biophys Acta Gen Subj 2024; 1868:130565. [PMID: 38244702 DOI: 10.1016/j.bbagen.2024.130565] [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: 12/05/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
N-glycosylation and proper processing of N-glycans are required for the function of membrane proteins including cell surface receptors. Fibroblast growth factor receptor (FGFR) is involved in a wide variety of biological processes including embryonic development, osteogenesis, angiogenesis, and cell proliferation. Human FGFR3 contains six potential N-glycosylation sites, however, the roles of glycosylation have not been elucidated. The site-specific profiles of N-glycans of the FGFR3 extracellular domain expressed and secreted by CHO-K1 cells were examined, and glycan occupancies and structures of four sites were determined. The results indicated that most sites were fully occupied by glycans, and the dominant populations were the complex type. By examining single N-glycan deletion mutants of FGFR3, it was found that N262Q mutation significantly increased the population with oligomannose-type N-glycans, which was localized in the endoplasmic reticulum. Protein stability assay suggested that fraction with oligomannose-type N-glycans in the N262Q mutant is more stable than those in the wild type and other mutants. Furthermore, it was found that ligand-independent phosphorylation was significantly upregulated in N262Q mutants with complex type N-glycans. The findings suggest that N-glycans on N262 of FGFR3 affect the intracellular localization and phosphorylation status of the receptor.
Collapse
Affiliation(s)
- Ukichiro Hashimoto
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naoki Fujitani
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuaki Uehara
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiromi Okamoto
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Saitou
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Fumie Ito
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shigeru Ariki
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Chemistry, Center for Medical Education, Sapporo Medical University, Japan
| | - Akiko Shiratsuchi
- Department of Chemistry, Center for Medical Education, Sapporo Medical University, Japan
| | - Yoshihiro Hasegawa
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo, Japan.
| |
Collapse
|
18
|
Truchan K, Osyczka AM. Noggin promotes osteogenesis in human adipose-derived mesenchymal stem cells via FGFR2/Src/Akt and ERK signaling pathway. Sci Rep 2024; 14:6724. [PMID: 38509118 PMCID: PMC10954655 DOI: 10.1038/s41598-024-56858-w] [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: 11/07/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The balance between Noggin and bone morphogenetic proteins (BMPs) is important during early development and skeletal regenerative therapies. Noggin binds BMPs in the extracellular space, thereby preventing BMP signaling. However, Noggin may affect cell response not necessarily through the modulation of BMP signaling, raising the possibility of direct Noggin signaling through yet unspecified receptors. Here we show that in osteogenic cultures of adipose-derived stem cells (ASCs), Noggin activates fibroblast growth factor receptors (FGFRs), Src/Akt and ERK kinases, and it stabilizes TAZ proteins in the presence of dexamethasone. Overall, this leads ASCs to increased expression of osteogenic markers and robust mineral deposition. Our results also indicate that Noggin can induce osteogenic genes expression in normal human bone marrow stem cells and alkaline phosphatase activity in normal human dental pulp stem cells. Besides, Noggin can specifically activate FGFR2 in osteosarcoma cells. We believe our findings open new research avenues to further explore the involvement of Noggin in cell fate modulation by FGFR2/Src/Akt/ERK signaling and potential applications of Noggin in bone regenerative therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| |
Collapse
|
19
|
Cheng YS, Taniguchi Y, Yunoki Y, Masai S, Nogi M, Doi H, Sekiguchi K, Nakagawa M. Simultaneous binding of bFGF to both FGFR and integrin maintains properties of primed human induced pluripotent stem cells. Regen Ther 2024; 25:113-127. [PMID: 38226057 PMCID: PMC10788407 DOI: 10.1016/j.reth.2023.12.008] [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: 10/03/2023] [Revised: 12/07/2023] [Accepted: 12/17/2023] [Indexed: 01/17/2024] Open
Abstract
Introduction Basic fibroblast growth factor (bFGF, FGF2) and integrin α6β1 are important for maintaining the pluripotency of human pluripotent stem cells (hPSCs). Although bFGF-integrin binding contributes to biofunctions in cancer cells, the relationship in hPSCs remains unclear. Methods To investigate the relationship between bFGF and integrin in human induced pluripotent stem cells (hiPSCs), we generated recombinant human bFGF wild-type and mutant proteins, that do not bind to integrin, FGFR, or both. We then cultured hiPSCs with these recombinant bFGF proteins. To evaluate the abilities of recombinant bFGF proteins in maintaining hPSC properties, pluripotent markers, ERK activity, and focal adhesion structure were analyzed through flow cytometry, immunofluorescence (IF), and immunoblotting (IB). Result We identified an interaction between bFGF and integrin α6β1 in vitro and in hiPSCs. The integrin non-binding mutant was incapable of inducing the hPSC properties, such as proliferation, ERK activity, and large focal adhesions at the edges of hiPSC colonies. Signaling induced by bFGF-FGFR binding was essential during the first 24 h after cell seeding for maintaining the properties of hPSCs, followed by a shift towards intracellular signaling via the bFGF-integrin interaction. The mixture of the two bFGF mutants also failed to maintain hPSC properties, indicating that bFGF binds to both FGFR and integrin. Conclusion Our study demonstrates that the integrin-bFGF-FGFR ternary complex maintains the properties of hPSCs via intracellular signaling, providing insights into the functional crosstalk between bFGF and integrins in hiPSCs.
Collapse
Affiliation(s)
- Yu-Shen Cheng
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Yukimasa Taniguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Yasuhiro Yunoki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Satomi Masai
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Mizuho Nogi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Hatsuki Doi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
20
|
Lolicato F, Steringer JP, Saleppico R, Beyer D, Fernandez-Sobaberas J, Unger S, Klein S, Riegerová P, Wegehingel S, Müller HM, Schmitt XJ, Kaptan S, Freund C, Hof M, Šachl R, Chlanda P, Vattulainen I, Nickel W. Disulfide bridge-dependent dimerization triggers FGF2 membrane translocation into the extracellular space. eLife 2024; 12:RP88579. [PMID: 38252473 PMCID: PMC10945597 DOI: 10.7554/elife.88579] [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] [Indexed: 01/23/2024] Open
Abstract
Fibroblast growth factor 2 (FGF2) exits cells by direct translocation across the plasma membrane, a type I pathway of unconventional protein secretion. This process is initiated by phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2)-dependent formation of highly dynamic FGF2 oligomers at the inner plasma membrane leaflet, inducing the formation of lipidic membrane pores. Cell surface heparan sulfate chains linked to glypican-1 (GPC1) capture FGF2 at the outer plasma membrane leaflet, completing FGF2 membrane translocation into the extracellular space. While the basic steps of this pathway are well understood, the molecular mechanism by which FGF2 oligomerizes on membrane surfaces remains unclear. In the current study, we demonstrate the initial step of this process to depend on C95-C95 disulfide-bridge-mediated FGF2 dimerization on membrane surfaces, producing the building blocks for higher FGF2 oligomers that drive the formation of membrane pores. We find FGF2 with a C95A substitution to be defective in oligomerization, pore formation, and membrane translocation. Consistently, we demonstrate a C95A variant of FGF2 to be characterized by a severe secretion phenotype. By contrast, while also important for efficient FGF2 secretion from cells, a second cysteine residue on the molecular surface of FGF2 (C77) is not involved in FGF2 oligomerization. Rather, we find C77 to be part of the interaction interface through which FGF2 binds to the α1 subunit of the Na,K-ATPase, the landing platform for FGF2 at the inner plasma membrane leaflet. Using cross-linking mass spectrometry, atomistic molecular dynamics simulations combined with a machine learning analysis and cryo-electron tomography, we propose a mechanism by which disulfide-bridged FGF2 dimers bind with high avidity to PI(4,5)P2 on membrane surfaces. We further propose a tight coupling between FGF2 secretion and the formation of ternary signaling complexes on cell surfaces, hypothesizing that C95-C95-bridged FGF2 dimers are functioning as the molecular units triggering autocrine and paracrine FGF2 signaling.
Collapse
Affiliation(s)
- Fabio Lolicato
- Heidelberg University Biochemistry CenterHeidelbergGermany
- Department of Physics, University of HelsinkiHelsinkiFinland
| | | | | | - Daniel Beyer
- Heidelberg University Biochemistry CenterHeidelbergGermany
| | | | | | - Steffen Klein
- Schaller Research Group, Department of Infectious Diseases-Virology, Heidelberg University HospitalHeidelbergGermany
| | - Petra Riegerová
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of SciencesPragueCzech Republic
| | | | | | - Xiao J Schmitt
- Institute for Chemistry and Biochemistry, Freie Universität BerlinBerlinGermany
| | - Shreyas Kaptan
- Department of Physics, University of HelsinkiHelsinkiFinland
| | - Christian Freund
- Institute for Chemistry and Biochemistry, Freie Universität BerlinBerlinGermany
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of SciencesPragueCzech Republic
| | - Radek Šachl
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of SciencesPragueCzech Republic
| | - Petr Chlanda
- Schaller Research Group, Department of Infectious Diseases-Virology, Heidelberg University HospitalHeidelbergGermany
| | | | - Walter Nickel
- Heidelberg University Biochemistry CenterHeidelbergGermany
| |
Collapse
|
21
|
Gutmann M, Reinhardt D, Seidensticker C, Raschig M, Hahn L, Moscaroli A, Behe M, Meinel L, Lühmann T. Matrix Metalloproteinase-Responsive Delivery of PEGylated Fibroblast Growth Factor 2. ACS Biomater Sci Eng 2024; 10:156-165. [PMID: 37988287 DOI: 10.1021/acsbiomaterials.3c01511] [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] [Indexed: 11/23/2023]
Abstract
Attachment of polyethylene glycol (PEG) chains is a common, well-studied, and Food and Drug Administration-approved method to address the pharmacokinetic challenges of therapeutic proteins. Occasionally, PEGylation impairs the activity of pharmacodynamics (PD). To overcome this problem, disease-relevant cleavable linkers between the polymer and the therapeutic protein can unleash full PD by de-PEGylating the protein at its target site. In this study, we engineered a matrix metalloproteinase (MMP)-responsive fibroblast growth factor 2 (FGF-2) mutant that was site-specifically extended with a PEG polymer chain. Using bioinspired strategies, the bioconjugate was designed to release the native protein at the desired structure/environment with preservation of the proliferative capacity in vitro on NIH3T3 cells. In vivo, hepatic exposure was diminished but not its renal distribution over time compared to unconjugated FGF-2. By releasing the growth factor from the PEG polymer in response to MMP cleavage, restored FGF-2 may enter hard-to-reach tissues and activate cell surface receptors or nuclear targets.
Collapse
Affiliation(s)
- Marcus Gutmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, DE-97074 Würzburg, Germany
| | - Debora Reinhardt
- Institute of Pharmacy and Food Chemistry, University of Würzburg, DE-97074 Würzburg, Germany
| | - Christian Seidensticker
- Medizinische Klinik und Poliklinik Für Innere Medizin II, Klinikum Rechts der Isar der TU München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Martina Raschig
- Institute of Pharmacy and Food Chemistry, University of Würzburg, DE-97074 Würzburg, Germany
| | - Lukas Hahn
- Institute of Pharmacy and Food Chemistry, University of Würzburg, DE-97074 Würzburg, Germany
| | - Alessandra Moscaroli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, DE-97074 Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), DE-97080 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, DE-97074 Würzburg, Germany
| |
Collapse
|
22
|
Gupta N, Kumar H, Gupta S, S M B, Saini K. A Concise Review on Natural Products and Their Derivatives for Breast Cancer Treatment. Chem Biodivers 2023; 20:e202300688. [PMID: 37431959 DOI: 10.1002/cbdv.202300688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/12/2023]
Abstract
Cancer is a leading cause of death worldwide. Among other cancers, breast cancer has been found to produce maximum number of cases in 2020. Different factors including geographical, genetic, hormonal, oral contraceptives and modern lifestyle could be responsible for the development of breast cancer and different pathways can be targeted for breast cancer treatment. The various conventional approaches used for the treatment of breast cancer including radiotherapy, chemotherapy, hormone and immunotherapy. But due to the side effects associated with these conventional treatments such as non-selectivity, multidrug resistance and bioavailability, there is a need for the development of better therapeutic agents for breast cancer treatment. Several natural products have been explored for breast cancer treatment. However, many of these natural products suffered from the limitations of poor water solubility and possess toxic side effects. To overcome these limitations, several structural analogs of natural products have been synthesized and possess potent anti-breast cancer effects with less side effects over their precursor molecules. In the present manuscript, we describe the pathogenesis of breast cancer, some potent natural products used in the treatment of breast cancer and their selected structural analogs possessing potent anti-breast cancer effects. Database such as Science direct, Pubmed and Google scholar were searched using keywords 'risk factors', 'screening methods','receptors', and 'natural products and derivatives', Registered clinical trials on selected natural products were also analyzed. Present study concludes that eight selected natural products and their derivatives possess wide potential to exhibit anti-breast cancer effects and could be explored further to develop better chemotherapeutic agents against breast cancer.
Collapse
Affiliation(s)
- Nidhi Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| | - Hitesh Kumar
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| | - Sumeet Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| | - Basavarajaiah S M
- PG Department of Chemistry, Vijaya College, RV Road, Bengaluru, 560004
| | - Kamal Saini
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| |
Collapse
|
23
|
Zhou Y, Sun S, Ling T, Chen Y, Zhou R, You Q. The role of fibroblast growth factor 18 in cancers: functions and signaling pathways. Front Oncol 2023; 13:1124520. [PMID: 37228502 PMCID: PMC10203589 DOI: 10.3389/fonc.2023.1124520] [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: 12/15/2022] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Fibroblast growth factor 18(FGF18) is a member of the fibroblast growth factor family (FGFs). FGF18 is a class of bioactive substances that can conduct biological signals, regulate cell growth, participate in tissue repair and other functions, and can promote the occurrence and development of different types of malignant tumors through various mechanisms. In this review, we focus on recent studies of FGF18 in the diagnosis, treatment, and prognosis of tumors in digestive, reproductive, urinary, respiratory, motor, and pediatric systems. These findings suggest that FGF18 may play an increasingly important role in the clinical evaluation of these malignancies. Overall, FGF18 can function as an important oncogene at different gene and protein levels, and can be used as a potential new therapeutic target and prognostic biomarker for these tumors.
Collapse
Affiliation(s)
- Yiming Zhou
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Sizheng Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Ling
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yongzhen Chen
- Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Rongzhong Zhou
- Department of Ophthalmology, Zaoyang First People’s Hosipital, Zaoyang, China
| | - Qiang You
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Freitag JS, Möser C, Belay R, Altattan B, Grasse N, Pothineni BK, Schnauß J, Smith DM. Integration of functional peptides into nucleic acid-based nanostructures. NANOSCALE 2023; 15:7608-7624. [PMID: 37042085 DOI: 10.1039/d2nr05429a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In many applications such as diagnostics and therapy development, small peptide fragments consisting of only a few amino acids are often attractive alternatives to bulky proteins. This is due to factors such as the ease of scalable chemical synthesis and numerous methods for their discovery. One drawback of using peptides is that their activity can often be negatively impacted by the lack of a rigid, 3D stabilizing structure provided by the rest of the protein. In many cases, this can be alleviated by different methods of rational templating onto nanomaterials, which provides additional possibilities to use concepts of multivalence or rational nano-engineering to enhance or even create new types of function or structure. In recent years, nanostructures made from the self-assembly of DNA strands have been used as scaffolds to create functional arrangements of peptides, often leading to greatly enhanced biological activity or new material properties. This review will give an overview of nano-templating approaches based on the combination of DNA nanotechnology and peptides. This will include both bioengineering strategies to control interactions with cells or other biological systems, as well as examples where the combination of DNA and peptides has been leveraged for the rational design of new functional materials.
Collapse
Affiliation(s)
- Jessica S Freitag
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Christin Möser
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Robel Belay
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Basma Altattan
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Nico Grasse
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | | | - Jörg Schnauß
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany
- Unconventional Computing Lab, UWE, Bristol, BS16 1QY, UK
| | - David M Smith
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig Medical Faculty, 04103 Leipzig, Germany
| |
Collapse
|
25
|
Pagano K, Listro R, Linciano P, Rossi D, Longhi E, Taraboletti G, Molinari H, Collina S, Ragona L. Identification of a novel extracellular inhibitor of FGF2/FGFR signaling axis by combined virtual screening and NMR spectroscopy approach. Bioorg Chem 2023; 136:106529. [PMID: 37084585 DOI: 10.1016/j.bioorg.2023.106529] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023]
Abstract
The aberrant activation of the fibroblast growth factor 2 (FGF2)/fibroblast growth factor receptor (FGFR) signalling pathway drives severe pathologies, including cancer development and angiogenesis-driven pathologies. The perturbation of the FGF2/FGFR axis via extracellular allosteric small inhibitors is a promising strategy for developing FGFR inhibitors with improved safety and efficacy for cancer treatment. We have previously investigated the role of new extracellular inhibitors, such as rosmarinic acid (RA), which bind the FGFR-D2 domain and directly compete with FGF2 for the same binding site, enabling the disruption of the functional FGF2/FGFR interaction. To select ligands for the previously identified FGF2/FGFR RA binding site, NMR data-driven virtual screening has been performed on an in-house library of non-commercial small molecules and metabolites. A novel drug-like compound, a resorcinol derivative named RBA4 has been identified. NMR interaction studies demonstrate that RBA4 binds the FGF2/FGFR complex, in agreement with docking prediction. Residue-level NMR perturbations analysis highlights that the mode of action of RBA4 is similar to RA in terms of its ability to target the FGF2/FGFR-D2 complex, inducing perturbations on both proteins and triggering complex dissociation. Biological assays proved that RBA4 inhibited FGF2 proliferative activity at a level comparable to the previously reported natural product, RA. Identification of RBA4 chemical groups involved in direct interactions represents a starting point for further optimization of drug-like extracellular inhibitors with improved activity.
Collapse
Affiliation(s)
- Katiuscia Pagano
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), Consiglio Nazionale delle Ricerche, via Corti 12, 20133 Milano, Italy
| | - Roberta Listro
- University of Pavia, Department of Drug Sciences, Via Taramelli 12, 27100 Pavia, Italy
| | - Pasquale Linciano
- University of Pavia, Department of Drug Sciences, Via Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- University of Pavia, Department of Drug Sciences, Via Taramelli 12, 27100 Pavia, Italy.
| | - Elisa Longhi
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche, Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Giulia Taraboletti
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche, Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Henriette Molinari
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), Consiglio Nazionale delle Ricerche, via Corti 12, 20133 Milano, Italy
| | - Simona Collina
- University of Pavia, Department of Drug Sciences, Via Taramelli 12, 27100 Pavia, Italy
| | - Laura Ragona
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" (SCITEC), Consiglio Nazionale delle Ricerche, via Corti 12, 20133 Milano, Italy.
| |
Collapse
|
26
|
Li H, Chen X, Zuo Z, Wang J, Guo Y. Identification and Characterization of Peptides from Bovine Collagen Hydrolysates that Promote Myogenic Cell Proliferation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4876-4889. [PMID: 36917229 DOI: 10.1021/acs.jafc.2c08929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
In this study, bovine collagen hydrolysate was purified via a series of chromatograms, and the peptides with the highest activity for promoting myoblast proliferation were identified by LC-MS-MS. It was demonstrated that the peptide GDAGPPGPAGPAGPPGPIG (hydroxylation) could promote C2C12 proliferation (+18.5% ± 0.04, P < 0.05). The certain peptide was capable of regulating the myogenic cell cycle and inhibiting myogenic cell apoptosis. By combining molecular docking, quantitative real-time PCR, and metabonomics, we suggested that the peptide GDAGPPGPAGPAGPPGPIG (hydroxylation) might bind to FGFR1 and affect the expression of genes downstream of FGFR1 and influence protein synthesis to promote myoblast proliferation. The above results showed that the peptides isolated in this study have the potential to alleviate sarcopenia in the elderly.
Collapse
Affiliation(s)
- Hanfeng Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| | - Xin Chen
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| | - Zhijie Zuo
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| | - Jianing Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yanchuan Guo
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| |
Collapse
|
27
|
Mendelian inheritance revisited: dominance and recessiveness in medical genetics. Nat Rev Genet 2023:10.1038/s41576-023-00574-0. [PMID: 36806206 DOI: 10.1038/s41576-023-00574-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2022] [Indexed: 02/22/2023]
Abstract
Understanding the consequences of genotype for phenotype (which ranges from molecule-level effects to whole-organism traits) is at the core of genetic diagnostics in medicine. Many measures of the deleteriousness of individual alleles exist, but these have limitations for predicting the clinical consequences. Various mechanisms can protect the organism from the adverse effects of functional variants, especially when the variant is paired with a wild type allele. Understanding why some alleles are harmful in the heterozygous state - representing dominant inheritance - but others only with the biallelic presence of pathogenic variants - representing recessive inheritance - is particularly important when faced with the deluge of rare genetic alterations identified by high throughput DNA sequencing. Both awareness of the specific quantitative and/or qualitative effects of individual variants and the elucidation of allelic and non-allelic interactions are essential to optimize genetic diagnosis and counselling.
Collapse
|
28
|
Xu Q, Dunbrack R. The protein common assembly database (ProtCAD)-a comprehensive structural resource of protein complexes. Nucleic Acids Res 2023; 51:D466-D478. [PMID: 36300618 PMCID: PMC9825537 DOI: 10.1093/nar/gkac937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 01/29/2023] Open
Abstract
Proteins often act through oligomeric interactions with other proteins. X-ray crystallography and cryo-electron microscopy provide detailed information on the structures of biological assemblies, defined as the most likely biologically relevant structures derived from experimental data. In crystal structures, the most relevant assembly may be ambiguously determined, since multiple assemblies observed in the crystal lattice may be plausible. It is estimated that 10-15% of PDB entries may have incorrect or ambiguous assembly annotations. Accurate assemblies are required for understanding functional data and training of deep learning methods for predicting assembly structures. As with any other kind of biological data, replication via multiple independent experiments provides important validation for the determination of biological assembly structures. Here we present the Protein Common Assembly Database (ProtCAD), which presents clusters of protein assembly structures observed in independent structure determinations of homologous proteins in the Protein Data Bank (PDB). ProtCAD is searchable by PDB entry, UniProt identifiers, or Pfam domain designations and provides downloads of coordinate files, PyMol scripts, and publicly available assembly annotations for each cluster of assemblies. About 60% of PDB entries contain assemblies in clusters of at least 2 independent experiments. All clusters and coordinates are available on ProtCAD web site (http://dunbrack2.fccc.edu/protcad).
Collapse
Affiliation(s)
- Qifang Xu
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Roland L Dunbrack
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
29
|
Hoellerbauer P, Biery MC, Arora S, Rao Y, Girard EJ, Mitchell K, Dighe P, Kufeld M, Kuppers DA, Herman JA, Holland EC, Soroceanu L, Vitanza NA, Olson JM, Pritchard JR, Paddison PJ. Functional genomic analysis of adult and pediatric brain tumor isolates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522885. [PMID: 36711964 PMCID: PMC9881972 DOI: 10.1101/2023.01.05.522885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Adult and pediatric tumors display stark differences in their mutation spectra and chromosome alterations. Here, we attempted to identify common and unique gene dependencies and their associated biomarkers among adult and pediatric tumor isolates using functional genetic lethal screens and computational modeling. Methods We performed CRISRP-Cas9 lethality screens in two adult glioblastoma (GBM) tumor isolates and five pediatric brain tumor isolates representing atypical teratoid rhabdoid tumors (ATRT), diffuse intrinsic pontine glioma, GBM, and medulloblastoma. We then integrated the screen results with machine learning-based gene-dependency models generated from data from >900 cancer cell lines. Results We found that >50% of candidate dependencies of 280 identified were shared between adult GBM tumors and individual pediatric tumor isolates. 68% of screen hits were found as nodes in our network models, along with shared and tumor-specific predictors of gene dependencies. We investigated network predictors associated with ADAR, EFR3A, FGFR1 (pediatric-specific), and SMARCC2 (ATRT-specific) gene dependency among our tumor isolates. Conclusions The results suggest that, despite harboring disparate genomic signatures, adult and pediatric tumor isolates share a preponderance of genetic dependences. Further, combining data from primary brain tumor lethality screens with large cancer cell line datasets produced valuable insights into biomarkers of gene dependency, even for rare cancers. Importance of the Study Our results demonstrate that large cancer cell lines data sets can be computationally mined to identify known and novel gene dependency relationships in adult and pediatric human brain tumor isolates. Gene dependency networks and lethality screen results represent a key resource for neuro-oncology and cancer research communities. We also highlight some of the challenges and limitations of this approach.
Collapse
Affiliation(s)
- Pia Hoellerbauer
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA USA
| | - Matt C Biery
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sonali Arora
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Yiyun Rao
- Huck Institute for the Life Sciences, Pennsylvania State University, State College, PA, USA
| | - Emily J Girard
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Kelly Mitchell
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Pratiksha Dighe
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Megan Kufeld
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Daniel A Kuppers
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Jacob A Herman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Eric C Holland
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Liliana Soroceanu
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Nicholas A Vitanza
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Justin R Pritchard
- Huck Institute for the Life Sciences, Pennsylvania State University, State College, PA, USA
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA USA
| |
Collapse
|
30
|
Chan J, Chan J, Shao L, Stawicki SS, Pham VC, Akita RW, Hafner M, Crocker L, Yu K, Koerber JT, Schaefer G, Comps-Agrar L. Systematic pharmacological analysis of agonistic and antagonistic fibroblast growth factor receptor 1 MAbs reveals a similar unique mode of action. J Biol Chem 2023; 299:102729. [PMID: 36410439 PMCID: PMC9758440 DOI: 10.1016/j.jbc.2022.102729] [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: 08/19/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/21/2022] Open
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is a receptor tyrosine kinase that plays a major role in developmental processes and metabolism. The dysregulation of FGFR1 through genetic aberrations leads to skeletal and metabolic diseases as well as cancer. For this reason, FGFR1 is a promising therapeutic target, yet a very challenging one due to potential on-target toxicity. More puzzling is that both agonistic and antagonistic FGFR1 antibodies are reported to exhibit similar toxicity profiles in vivo, namely weight loss. In this study, we aimed to assess and compare the mechanism of action of these molecules to better understand this apparent contradiction. By systematically comparing the binding of these antibodies and the activation or the inhibition of the major FGFR1 signaling events, we demonstrated that the molecules displayed similar properties and can behave either as an agonist or antagonist depending on the presence or the absence of the endogenous ligand. We further demonstrated that these findings translated in xenografts mice models. In addition, using time-resolved FRET and mass spectrometry analysis, we showed a functionally distinct FGFR1 active conformation in the presence of an antibody that preferentially activates the FGFR substrate 2 (FRS2)-dependent signaling pathway, demonstrating that modulating the geometry of a FGFR1 dimer can effectively change the signaling outputs and ultimately the activity of the molecule in preclinical studies. Altogether, our results highlighted how bivalent antibodies can exhibit both agonistic and antagonistic activities and have implications for targeting other receptor tyrosine kinases with antibodies.
Collapse
Affiliation(s)
- Jocelyn Chan
- Department of Molecular Oncology, Genentech Inc, South San Francisco, California, USA
| | - Joyce Chan
- Department of Biochemical and Cellular Pharmacology, Genentech Inc, South San Francisco, USA
| | - Lily Shao
- Department of Molecular Oncology, Genentech Inc, South San Francisco, California, USA
| | - Scott S Stawicki
- Department of Antibody Engineering, Genentech Inc, South San Francisco, California, USA
| | - Victoria C Pham
- Department of Microchemistry, Proteomics, Lipidomics and NGS, Genentech Inc, South San Francisco, California, USA
| | - Rob W Akita
- Department of Molecular Oncology, Genentech Inc, South San Francisco, California, USA
| | - Marc Hafner
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA
| | - Lisa Crocker
- Department of Translational Oncology, Genentech Inc, South San Francisco, California, USA
| | - Kebing Yu
- Department of Microchemistry, Proteomics, Lipidomics and NGS, Genentech Inc, South San Francisco, California, USA
| | - James T Koerber
- Department of Antibody Engineering, Genentech Inc, South San Francisco, California, USA
| | - Gabriele Schaefer
- Department of Molecular Oncology, Genentech Inc, South San Francisco, California, USA.
| | - Laetitia Comps-Agrar
- Department of Biochemical and Cellular Pharmacology, Genentech Inc, South San Francisco, USA.
| |
Collapse
|
31
|
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
|
32
|
Pagano K, Longhi E, Molinari H, Taraboletti G, Ragona L. Inhibition of FGFR Signaling by Targeting FGF/FGFR Extracellular Interactions: Towards the Comprehension of the Molecular Mechanism through NMR Approaches. Int J Mol Sci 2022; 23:ijms231810860. [PMID: 36142770 PMCID: PMC9503799 DOI: 10.3390/ijms231810860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
NMR-based approaches play a pivotal role in providing insight into molecular recognition mechanisms, affording the required atomic-level description and enabling the identification of promising inhibitors of protein–protein interactions. The aberrant activation of the fibroblast growth factor 2 (FGF2)/fibroblast growth factor receptor (FGFR) signaling pathway drives several pathologies, including cancer development, metastasis formation, resistance to therapy, angiogenesis-driven pathologies, vascular diseases, and viral infections. Most FGFR inhibitors targeting the intracellular ATP binding pocket of FGFR have adverse effects, such as limited specificity and relevant toxicity. A viable alternative is represented by targeting the FGF/FGFR extracellular interactions. We previously identified a few small-molecule inhibitors acting extracellularly, targeting FGFR or FGF. We have now built a small library of natural and synthetic molecules that potentially act as inhibitors of FGF2/FGFR interactions to improve our understanding of the molecular mechanisms of inhibitory activity. Here, we provide a comparative analysis of the interaction mode of small molecules with the FGF2/FGFR complex and the single protein domains. DOSY and residue-level NMR analysis afforded insights into the capability of the potential inhibitors to destabilize complex formation, highlighting different mechanisms of inhibition of FGF2-induced cell proliferation.
Collapse
Affiliation(s)
- Katiuscia Pagano
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” (SCITEC), via Corti 12, 20133 Milano, Italy
- Correspondence: (K.P.); (L.R.)
| | - Elisa Longhi
- Laboratory of Tumour Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Henriette Molinari
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” (SCITEC), via Corti 12, 20133 Milano, Italy
| | - Giulia Taraboletti
- Laboratory of Tumour Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Laura Ragona
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” (SCITEC), via Corti 12, 20133 Milano, Italy
- Correspondence: (K.P.); (L.R.)
| |
Collapse
|
33
|
Du X, McManus DP, Fogarty CE, Jones MK, You H. Schistosoma mansoni Fibroblast Growth Factor Receptor A Orchestrates Multiple Functions in Schistosome Biology and in the Host-Parasite Interplay. Front Immunol 2022; 13:868077. [PMID: 35812433 PMCID: PMC9257043 DOI: 10.3389/fimmu.2022.868077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022] Open
Abstract
Stem cells play significant roles in driving the complex life cycle of Schistosoma mansoni. Fibroblast growth factor (FGF) receptor A (SmFGFRA) is essential for maintaining the integrity of schistosome stem cells. Using immunolocalization, we demonstrated that SmFGFRA was distributed abundantly in germinal/stem cells of different S. mansoni life stages including eggs, miracidia, cercariae, schistosomula and adult worms. Indeed, SmFGFRA was also localized amply in embryonic cells and in the perinuclear region of immature eggs; von Lichtenberg's layer and the neural mass of mature eggs; the ciliated surface and neural mass of miracidia; the tegument cytosol of cercariae, schistosomula and adult worms; and was present in abundance in the testis and vitellaria of adult worms of S. mansoni. The distribution pattern of SmFGFRA illustrates the importance of this molecule in maintaining stem cells, development of the nervous and reproductive system of schistosomes, and in the host-parasite interplay. We showed SmFGFRA can bind human FGFs, activating the mitogen activated protein kinase (MAPK) pathway of adult worms in vitro. Inhibition of FGF signaling by the specific tyrosine kinase inhibitor BIBF 1120 significantly reduced egg hatching ability and affected the behavior of miracidia hatched from the treated eggs, emphasizing the importance of FGF signaling in driving the life cycle of S. mansoni. Our findings provide increased understanding of the complex schistosome life cycle and host-parasite interactions, indicating components of the FGF signaling pathway may represent promising targets for developing new interventions against schistosomiasis.
Collapse
Affiliation(s)
- Xiaofeng Du
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Donald P. McManus
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Conor E. Fogarty
- Genecology Research Centre, University of the Sunshine Coast, Brisbane, QLD, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
| | - Hong You
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Gasser E, Sancar G, Downes M, Evans RM. Metabolic Messengers: fibroblast growth factor 1. Nat Metab 2022; 4:663-671. [PMID: 35681108 PMCID: PMC9624216 DOI: 10.1038/s42255-022-00580-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/15/2022] [Accepted: 04/27/2022] [Indexed: 11/09/2022]
Abstract
While fibroblast growth factor (FGF) 1 is expressed in multiple tissues, only adipose-derived and brain FGF1 have been implicated in the regulation of metabolism. Adipose FGF1 production is upregulated in response to dietary stress and is essential for adipose tissue plasticity in these conditions. Similarly, in the brain, FGF1 secretion into the ventricular space and the adjacent parenchyma is increased after a hypercaloric challenge induced by either feeding or glucose infusion. Potent anorexigenic properties have been ascribed to both peripheral and centrally injected FGF1. The ability of recombinant FGF1 and variants with reduced mitogenicity to lower glucose, suppress adipose lipolysis and promote insulin sensitization elevates their potential as candidates in the treatment of type 2 diabetes mellitus and associated comorbidities. Here, we provide an overview of the known metabolic functions of endogenous FGF1 and discuss its therapeutic potential, distinguishing between peripherally or centrally administered FGF1.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Gencer Sancar
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
35
|
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
|
36
|
Cao L, Coventry B, Goreshnik I, Huang B, Sheffler W, Park JS, Jude KM, Marković I, Kadam RU, Verschueren KHG, Verstraete K, Walsh STR, Bennett N, Phal A, Yang A, Kozodoy L, DeWitt M, Picton L, Miller L, Strauch EM, DeBouver ND, Pires A, Bera AK, Halabiya S, Hammerson B, Yang W, Bernard S, Stewart L, Wilson IA, Ruohola-Baker H, Schlessinger J, Lee S, Savvides SN, Garcia KC, Baker D. Design of protein-binding proteins from the target structure alone. Nature 2022; 605:551-560. [PMID: 35332283 PMCID: PMC9117152 DOI: 10.1038/s41586-022-04654-9] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/15/2022] [Indexed: 12/03/2022]
Abstract
The design of proteins that bind to a specific site on the surface of a target protein using no information other than the three-dimensional structure of the target remains a challenge1-5. Here we describe a general solution to this problem that starts with a broad exploration of the vast space of possible binding modes to a selected region of a protein surface, and then intensifies the search in the vicinity of the most promising binding modes. We demonstrate the broad applicability of this approach through the de novo design of binding proteins to 12 diverse protein targets with different shapes and surface properties. Biophysical characterization shows that the binders, which are all smaller than 65 amino acids, are hyperstable and, following experimental optimization, bind their targets with nanomolar to picomolar affinities. We succeeded in solving crystal structures of five of the binder-target complexes, and all five closely match the corresponding computational design models. Experimental data on nearly half a million computational designs and hundreds of thousands of point mutants provide detailed feedback on the strengths and limitations of the method and of our current understanding of protein-protein interactions, and should guide improvements of both. Our approach enables the targeted design of binders to sites of interest on a wide variety of proteins for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Longxing Cao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Buwei Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - William Sheffler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Joon Sung Park
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin M Jude
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Iva Marković
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Rameshwar U Kadam
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Koen H G Verschueren
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Kenneth Verstraete
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Scott Thomas Russell Walsh
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
- J.A.M.E.S. Farm, Clarksville, MD, USA
| | - Nathaniel Bennett
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | - Ashish Phal
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Aerin Yang
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa Kozodoy
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Michelle DeWitt
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lora Picton
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauren Miller
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Eva-Maria Strauch
- Deptartment of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Nicholas D DeBouver
- UCB Pharma, Bainbridge Island, WA, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Allison Pires
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
- Seattle Children's Center for Global Infectious Disease Research, Seattle, WA, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samer Halabiya
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Bradley Hammerson
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA, USA
| | - Wei Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Steffen Bernard
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Lance Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Joseph Schlessinger
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Sangwon Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Savvas N Savvides
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
37
|
Unconventional secretion mediated by direct protein self-translocation across the plasma membranes of mammalian cells. Trends Biochem Sci 2022; 47:699-709. [PMID: 35490075 DOI: 10.1016/j.tibs.2022.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 12/17/2022]
Abstract
In recent years, a surprisingly complex picture emerged about endoplasmic reticulum (ER)/Golgi-independent secretory pathways, and several routes have been discovered that differ with regard to their molecular mechanisms and machineries. Fibroblast growth factor 2 (FGF2) is secreted by a pathway of unconventional protein secretion (UPS) that is based on direct self-translocation across the plasma membrane. Building on previous research, a component of this process has been identified to be glypican-1 (GPC1), a GPI-anchored heparan sulfate proteoglycan located on cell surfaces. These findings not only shed light on the molecular mechanism underlying this process but also reveal an intimate relationship between FGF2 and GPC1 that might be of critical relevance for the prominent roles they both have in tumor progression and metastasis.
Collapse
|
38
|
Lolicato F, Nickel W. A Role for Liquid-Ordered Plasma Membrane Nanodomains Coordinating the Unconventional Secretory Pathway of Fibroblast Growth Factor 2? Front Cell Dev Biol 2022; 10:864257. [PMID: 35433697 PMCID: PMC9010882 DOI: 10.3389/fcell.2022.864257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022] Open
Abstract
Fibroblast growth factor 2 (FGF2) is a tumor cell survival factor that belongs to a subgroup of extracellular proteins lacking N-terminal signal peptides. Whereas this phenomenon was already recognized in the early 1990s, detailed insights into the molecular mechanisms underlying alternative pathways of protein secretion from eukaryotic cells were obtained only recently. Today, we know about a number of alternative secretory mechanisms, collectively termed unconventional protein secretion (UPS). FGF2 belongs to a subgroup of cargo proteins secreted by direct translocation across the plasma membrane. This feature has been classified as type I UPS and is shared with other unconventionally secreted proteins, such as HIV-Tat and Tau. FGF2 translocation across the membrane is initiated through sequential interactions with the Na,K-ATPase, Tec kinase, and phosphoinositide PI(4,5)P2 at the inner plasma membrane leaflet. Whereas the first two are auxiliary factors of this pathway, the interaction of FGF2 with PI(4,5)P2 triggers the core mechanism of FGF2 membrane translocation. It is based on a lipidic membrane pore that is formed by PI(4,5)P2-induced oligomerization of FGF2. Membrane-inserted FGF2 oligomers are recognized as translocation intermediates that are resolved at the outer plasma membrane leaflet by glypican-1, a heparan sulfate proteoglycan that captures and disassembles FGF2 oligomers on cell surfaces. Here, we discuss recent findings suggesting the molecular machinery mediating FGF2 membrane translocation to be highly organized in liquid-ordered plasma membrane nanodomains, the core process underlying this unusual pathway of protein secretion.
Collapse
Affiliation(s)
- Fabio Lolicato
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Walter Nickel
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| |
Collapse
|
39
|
Exploring the FGF/FGFR System in Ocular Tumors: New Insights and Perspectives. Int J Mol Sci 2022; 23:ijms23073835. [PMID: 35409195 PMCID: PMC8998873 DOI: 10.3390/ijms23073835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022] Open
Abstract
Ocular tumors are a family of rare neoplasms that develop in the eye. Depending on the type of cancer, they mainly originate from cells localized within the retina, the uvea, or the vitreous. Even though current treatments (e.g., radiotherapy, transpupillary thermotherapy, cryotherapy, chemotherapy, local resection, or enucleation) achieve the control of the local tumor in the majority of treated cases, a significant percentage of patients develop metastatic disease. In recent years, new targeting therapies and immuno-therapeutic approaches have been evaluated. Nevertheless, the search for novel targets and players is eagerly required to prevent and control tumor growth and metastasis dissemination. The fibroblast growth factor (FGF)/FGF receptor (FGFR) system consists of a family of proteins involved in a variety of physiological and pathological processes, including cancer. Indeed, tumor and stroma activation of the FGF/FGFR system plays a relevant role in tumor growth, invasion, and resistance, as well as in angiogenesis and dissemination. To date, scattered pieces of literature report that FGFs and FGFRs are expressed by a significant subset of primary eye cancers, where they play relevant and pleiotropic roles. In this review, we provide an up-to-date description of the relevant roles played by the FGF/FGFR system in ocular tumors and speculate on its possible prognostic and therapeutic exploitation.
Collapse
|
40
|
Improvement of FGF7 Thermal Stability by Introduction of Mutations in Close Vicinity to Disulfide Bond and Surface Salt Bridge. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10394-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AbstractFibroblast Growth Factor 7 (FGF7), a growth factor specific to epithelial cells, has attracted attention as a therapeutic protein. However, FGF7 has a limitation in its use due to low protein stability. Here, the mutations were designed to increase the stability of FGF7 by analyzing its 3D structure and sequence of other FGFs. Palifermin, N-terminal truncated FGF7 is known to have improved stability and was used as control protein in our study. The K126 and K178 were substituted into glutamate to form salt bridge with the neighboring residue R175 respectively and A120C mutation was introduced in close vicinity to disulfide bond between C133 and C137. The data of Circular Dichroism (CD) showed that all mutant proteins tested had higher Tm value than Palifermin and Tm of A120C/K126E/K178E FGF7 mutant protein was 15.24 °C higher than that of Palifermin. The results of cell proliferation activity and soluble protein analyzed by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) after 37 °C or 45 °C incubation exhibited that the stability of A120C mutant protein and A120C-including mutant proteins was improved. These results suggest that the mutation of amino acid in close vicinity to disulfide bond and the salt bridge at the surface of FGF7 enhanced thermal stability and make FGF7 more useful for pharmaceutical and cosmetical application.
Collapse
|
41
|
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
|
42
|
Elangovan M, Ka J, Pak B, Choi W, Oh SR, Jin SW, Yoo YJ. Ubiquitin-conjugating enzyme V variant 1 enables cellular responses toward fibroblast growth factor signaling in endothelium. FASEB J 2021; 36:e22103. [PMID: 34921695 DOI: 10.1096/fj.202100808rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022]
Abstract
Ubiquitination has been shown to provide an essential regulatory role in modulating the duration and amplitude of the signaling activity in angiogenesis. While successive enzymatic reactions mediated by three distinct types of enzymes commonly known as E1, E2, and E3 are required for ubiquitination, the role of E3s which govern the final step of ubiquitination has been extensively analyzed in angiogenesis. In contrast, the role of E2s, which determine the context and functional consequences of ubiquitination, remains largely unknown with respect to angiogenesis. To better elucidate the role of E2s in modulating endothelial behaviors during angiogenesis, we first systematically analyze the expression pattern of E2s in endothelial cells (ECs) using previously published scRNA-seq data and identify ubiquitin-conjugating enzyme variant 1 (UBE2V1), an unconventional E2 without innate catalytic activity, as one of the most abundantly expressed E2s in ECs. While ubiquitously expressed in diverse cell types, abrogation of UBE2V1 significantly impairs proliferation and viability of human umbilical vein endothelial cells (HUVECs) without affecting other cell types, suggesting that UBE2V1 is likely to possess nonredundant functions in ECs. Consistent with this idea, UBE2V1 appears to be critical for morphogenesis and migration of ECs during angiogenesis. Interestingly, we find that UBE2V1 is essential for fibroblast growth factor 2 (FGF2)-induced angiogenesis, but appears to have minor effects on vascular endothelial growth factor-A-induced angiogenesis in vitro as well as in vivo. Therefore, it seems that UBE2V1 could enable ECs to distinguish two related yet distinct angiogenic cues. Mechanistically, we show that UBE2V1 promotes ubiquitination of MEK kinase 1, a key mediator of FGF2 signaling, to enhance phosphorylation of extracellular signal-regulated kinase 1/2 in HUVECs. Taken together, our results illustrate the unique role of UBE2V1 as a key modulator for angiogenic behaviors in ECs.
Collapse
Affiliation(s)
- Muthukumar Elangovan
- Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.,School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jun Ka
- Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.,School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Boryeong Pak
- Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.,School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Woosoung Choi
- Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.,School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Se-Ra Oh
- Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.,School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Suk-Won Jin
- Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.,School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yung Joon Yoo
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| |
Collapse
|
43
|
Hoshiyama J, Okada Y, Hayata Y, Eguchi A, Ueki R, Sando S. Characterization of a DNA Aptamer with High Specificity toward Fibroblast Growth Factor Receptor 1. CHEM LETT 2021. [DOI: 10.1246/cl.210505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Junya Hoshiyama
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yuga Okada
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yuri Hayata
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akihiro Eguchi
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ryosuke Ueki
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
44
|
Chioni AM, Grose RP. Biological Significance and Targeting of the FGFR Axis in Cancer. Cancers (Basel) 2021; 13:5681. [PMID: 34830836 PMCID: PMC8616401 DOI: 10.3390/cancers13225681] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
The pleiotropic effects of fibroblast growth factors (FGFs), the widespread expression of all seven signalling FGF receptors (FGFRs) throughout the body, and the dramatic phenotypes shown by many FGF/R knockout mice, highlight the diversity, complexity and functional importance of FGFR signalling. The FGF/R axis is critical during normal tissue development, homeostasis and repair. Therefore, it is not surprising that substantial evidence also pinpoints the involvement of aberrant FGFR signalling in disease, including tumourigenesis. FGFR aberrations in cancer include mutations, gene fusions, and amplifications as well as corrupted autocrine/paracrine loops. Indeed, many clinical trials on cancer are focusing on targeting the FGF/FGFR axis, using selective FGFR inhibitors, nonselective FGFR tyrosine kinase inhibitors, ligand traps, and monoclonal antibodies and some have already been approved for the treatment of cancer patients. The heterogeneous tumour microenvironment and complexity of FGFR signalling may be some of the factors responsible for the resistance or poor response to therapy with FGFR axis-directed therapeutic agents. In the present review we will focus on the structure and function of FGF(R)s, their common irregularities in cancer and the therapeutic value of targeting their function in cancer.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Richard P. Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK;
| |
Collapse
|
45
|
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
|
46
|
Aprajita, Sharma R. Comprehending fibroblast growth factor receptor like 1: Oncogene or tumor suppressor? Cancer Treat Res Commun 2021; 29:100472. [PMID: 34689016 DOI: 10.1016/j.ctarc.2021.100472] [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: 08/07/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/16/2022]
Abstract
Fibroblast Growth Factor Receptor Like 1 (FGFRL1) signaling has crucial role in a multitude of processes during genetic diseases, embryonic development and various types of cancer. Due to its partial structural similarity with its classical Fibroblast Growth Factor Receptor [FGFR] counterparts and lack of tyrosine kinase domain, FGFRL1 was thought to work as a decoy receptor in FGF/FGFR signaling. Later on, growing number evidences showed that expression of FGFRL1 affects major pathways like ERK1/2, Akt and others, which are dysfunctional in a wide range of human cancers. In this review, we provide an overview of the current understanding of FGFRL1 and its roles in cell differentiation, adhesion and proliferation pathways . Overexpression of FGFRL1 might lead to tumor progression and invasion. In this context, inhibitors for FGFRL1 might have therapeutic benefits in human cancer prognosis.
Collapse
Affiliation(s)
- Aprajita
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Dwarka, New Delhi, India
| | - Rinu Sharma
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Dwarka, New Delhi, India.
| |
Collapse
|
47
|
Karl K, Hristova K. Pondering the mechanism of receptor tyrosine kinase activation: The case for ligand-specific dimer microstate ensembles. Curr Opin Struct Biol 2021; 71:193-199. [PMID: 34399300 DOI: 10.1016/j.sbi.2021.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/04/2021] [Accepted: 07/04/2021] [Indexed: 10/20/2022]
Abstract
Receptor tyrosine kinases (RTKs) are single-pass membrane proteins that regulate cell growth, differentiation, motility, and metabolism. Here, we review recent advancements in RTK structure determination and in the understanding of RTK activation. We argue that further progress in the field will necessitate new ways of thinking, and we introduce the concept that RTK dimers explore ensembles of microstates, each characterized by different kinase domain dimer conformations, but the same extracellular domain dimer structure. Many microstates are phosphorylation-competent and ensure the phosphorylation of one specific tyrosine. The prevalence of each microstate correlates with its stability. A switch in ligand will lead to a switch in the extracellular domain configuration and to a subsequent switch in the ensemble of microstates. This model can explain how different ligands produce specific phosphorylation patterns, how receptor overexpression leads to enhanced signaling even in the absence of activating ligands, and why RTK kinase domain structures have remained unresolved in cryogenic electron microscopy studies.
Collapse
Affiliation(s)
- Kelly Karl
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218 USA
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218 USA.
| |
Collapse
|
48
|
Nita A, Abraham SP, Krejci P, Bosakova M. Oncogenic FGFR Fusions Produce Centrosome and Cilia Defects by Ectopic Signaling. Cells 2021; 10:1445. [PMID: 34207779 PMCID: PMC8227969 DOI: 10.3390/cells10061445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
A single primary cilium projects from most vertebrate cells to guide cell fate decisions. A growing list of signaling molecules is found to function through cilia and control ciliogenesis, including the fibroblast growth factor receptors (FGFR). Aberrant FGFR activity produces abnormal cilia with deregulated signaling, which contributes to pathogenesis of the FGFR-mediated genetic disorders. FGFR lesions are also found in cancer, raising a possibility of cilia involvement in the neoplastic transformation and tumor progression. Here, we focus on FGFR gene fusions, and discuss the possible mechanisms by which they function as oncogenic drivers. We show that a substantial portion of the FGFR fusion partners are proteins associated with the centrosome cycle, including organization of the mitotic spindle and ciliogenesis. The functions of centrosome proteins are often lost with the gene fusion, leading to haploinsufficiency that induces cilia loss and deregulated cell division. We speculate that this complements the ectopic FGFR activity and drives the FGFR fusion cancers.
Collapse
Affiliation(s)
- Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Sara P. Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
49
|
Gascon S, Jann J, Langlois-Blais C, Plourde M, Lavoie C, Faucheux N. Peptides Derived from Growth Factors to Treat Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22116071. [PMID: 34199883 PMCID: PMC8200100 DOI: 10.3390/ijms22116071] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by progressive neuron losses in memory-related brain structures. The classical features of AD are a dysregulation of the cholinergic system, the accumulation of amyloid plaques, and neurofibrillary tangles. Unfortunately, current treatments are unable to cure or even delay the progression of the disease. Therefore, new therapeutic strategies have emerged, such as the exogenous administration of neurotrophic factors (e.g., NGF and BDNF) that are deficient or dysregulated in AD. However, their low capacity to cross the blood-brain barrier and their exorbitant cost currently limit their use. To overcome these limitations, short peptides mimicking the binding receptor sites of these growth factors have been developed. Such peptides can target selective signaling pathways involved in neuron survival, differentiation, and/or maintenance. This review focuses on growth factors and their derived peptides as potential treatment for AD. It describes (1) the physiological functions of growth factors in the brain, their neuronal signaling pathways, and alteration in AD; (2) the strategies to develop peptides derived from growth factor and their capacity to mimic the role of native proteins; and (3) new advancements and potential in using these molecules as therapeutic treatments for AD, as well as their limitations.
Collapse
Affiliation(s)
- Suzanne Gascon
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
| | - Jessica Jann
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
| | - Chloé Langlois-Blais
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l’Estrie–Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1G 1B1, Canada;
- Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Christine Lavoie
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Institut de Pharmacologie de Sherbrooke, 3001 12th Avenue, N., Sherbrooke, QC J1H 5N4, Canada
- Correspondence: (C.L.); (N.F.); Tel.: +1-819-821-8000 (ext. 72732) (C.L.); +1-819-821-8000 (ext. 61343) (N.F.)
| | - Nathalie Faucheux
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, 2500 Boulevard Université, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (S.G.); (J.J.)
- Institut de Pharmacologie de Sherbrooke, 3001 12th Avenue, N., Sherbrooke, QC J1H 5N4, Canada
- Correspondence: (C.L.); (N.F.); Tel.: +1-819-821-8000 (ext. 72732) (C.L.); +1-819-821-8000 (ext. 61343) (N.F.)
| |
Collapse
|
50
|
Kinoshita N, Hashimoto Y, Yasue N, Suzuki M, Cristea IM, Ueno N. Mechanical Stress Regulates Epithelial Tissue Integrity and Stiffness through the FGFR/Erk2 Signaling Pathway during Embryogenesis. Cell Rep 2021; 30:3875-3888.e3. [PMID: 32187556 DOI: 10.1016/j.celrep.2020.02.074] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 01/31/2020] [Accepted: 02/19/2020] [Indexed: 12/22/2022] Open
Abstract
Physical forces generated by tissue-tissue interactions are a critical component of embryogenesis, aiding the formation of organs in a coordinated manner. In this study, using Xenopus laevis embryos and phosphoproteome analyses, we uncover the rapid activation of the mitogen-activated protein (MAP) kinase Erk2 upon stimulation with centrifugal, compression, or stretching force. We demonstrate that Erk2 induces the remodeling of cytoskeletal proteins, including F-actin, an embryonic cadherin C-cadherin, and the tight junction protein ZO-1. We show these force-dependent changes to be prerequisites for the enhancement of cellular junctions and tissue stiffening during early embryogenesis. Furthermore, Erk2 activation is FGFR1 dependent while not requiring fibroblast growth factor (FGF) ligands, suggesting that cell/tissue deformation triggers receptor activation in the absence of ligands. These findings establish previously unrecognized functions for mechanical forces in embryogenesis and reveal its underlying force-induced signaling pathways.
Collapse
Affiliation(s)
- Noriyuki Kinoshita
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Yutaka Hashimoto
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; International Research Collaboration Center, National Institutes of Natural Sciences, Tokyo 105-0001, Japan; Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Naoko Yasue
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Makoto Suzuki
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA.
| | - Naoto Ueno
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan; School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|