1
|
Swieton J, Miklosz J, Bielicka N, Frackiewicz A, Depczynski K, Stolarek M, Bonarek P, Kaminski K, Rozga P, Yusa SI, Gromotowicz-Poplawska A, Szczubialka K, Pawlak D, Mogielnicki A, Kalaska B. Synthesis, Biological Evaluation and Reversal of Sulfonated Di- and Triblock Copolymers as Novel Parenteral Anticoagulants. Adv Healthc Mater 2024; 13:e2402191. [PMID: 39370656 DOI: 10.1002/adhm.202402191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Despite targeting different coagulation cascade sites, all Food and Drug Administration-approved anticoagulants present an elevated risk of bleeding, including potentially life-threatening intracranial hemorrhage. Existing studies have not thoroughly investigated the efficacy and safety of sulfonate polymers in animal models and fully elucidate the precise mechanisms by which these polymers act. The activity and safety of sulfonated di- and triblock copolymers containing poly(sodium styrenesulfonate) (PSSS), poly(sodium 2-acrylamido-2-methylpropanesulfonate) (PAMPS), poly(ethylene glycol) (PEG), poly(sodium methacrylate) (PMAAS), poly(acrylic acid) (PAA), and poly(sodium 11-acrylamidoundecanoate) (PAaU) blocks are synthesized and assessed. PSSS-based copolymers exhibit greater anticoagulant activity than PAMPS-based ones. Their activity is mainly affected by the total concentration of sulfonate groups and molecular weight. PEG-containing copolymers demonstrate a better safety profile than PAA-containing ones. The selected copolymer PEG47-PSSS32 exhibits potent anticoagulant activity in rodents after subcutaneous and intravenous administration. Heparin Binding Copolymer (HBC) completely reverses the anticoagulant activity of polymer in rat and human plasma. No interaction with platelets is observed. Selected copolymer targets mainly factor XII and fibrinogen, and to a lesser extent factors X, IX, VIII, and II, suggesting potential application in blood-contacting biomaterials for anticoagulation purposes. Further studies are needed to explore its therapeutic applications fully.
Collapse
Affiliation(s)
- Justyna Swieton
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Joanna Miklosz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Natalia Bielicka
- Department of Biopharmacy and Radiopharmacy, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Aleksandra Frackiewicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Karol Depczynski
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Marta Stolarek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, prof. S. Lojasiewicza 11 St., Krakow, 30-348, Poland
| | - Piotr Bonarek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
| | - Kamil Kaminski
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
| | - Piotr Rozga
- Drug Discovery and Early Development Department, Adamed Pharma S.A., Pienkow, Mariana Adamkiewicza 6A St., Czosnow, 05-152, Poland
| | - Shin-Ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 167 Shosha, Himeji, 671-2280, Japan
| | - Anna Gromotowicz-Poplawska
- Department of Biopharmacy and Radiopharmacy, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Krzysztof Szczubialka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Andrzej Mogielnicki
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Bartlomiej Kalaska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| |
Collapse
|
2
|
Wang H, Wang J, Feng J, Rao Y, Xu Z, Zu J, Wang H, Zhang Z, Chen H. Artificial Extracellular Matrix Composed of Heparin-Mimicking Polymers for Efficient Anticoagulation and Promotion of Endothelial Cell Proliferation. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50142-50151. [PMID: 36302722 DOI: 10.1021/acsami.2c13892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Heparin-mimicking polymers have emerged as an alternative to heparin to construct effective and safe anticoagulant surfaces. However, the present heparin-mimicking polymers are usually limited to the combinations of glucose and sulfonic acid units, and the structure origin of their anticoagulant properties remains vague. Inspired by the structure of natural heparin, we synthesized a series of novel heparin-mimicking polymers (named GSAs) composed of three units, glucose, sulfonic acid, and carboxylic acid. Then, we constructed artificial extracellular matrices composed of GSAs and two typical cationic polymers, polyethyleneimine and chitosan, to investigate the anticoagulation and endothelialization of GSAs. By changing the ratio of the three units, their functions in the matrices were studied systematically. We found that an increase in the sulfonic acid content enhanced surface anticoagulant activity, an increase in glucose and sulfonic acid content promoted the proliferation of human umbilical vein vascular endothelial cells, and an increase in the carboxylic acid content inhibited the adherence of human umbilical vein vascular smooth muscle cells. This work uncovers the important role of the GSAs structure to the anticoagulation properties, which sheds new light on the design and preparation of heparin-mimicking polymers for practical engineering applications.
Collapse
Affiliation(s)
- Huanhuan Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - Jinghong Wang
- The SIP Biointerface Engineering Research Institute, Suzhou215123, P. R. China
| | - Jian Feng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - Yu Rao
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - ZiYing Xu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - JunYi Zu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - Huaguang Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - Zexin Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou215123, P. R. China
- The SIP Biointerface Engineering Research Institute, Suzhou215123, P. R. China
| |
Collapse
|
3
|
Novais A, Chatzopoulou E, Chaussain C, Gorin C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells 2021; 10:932. [PMID: 33920587 PMCID: PMC8073160 DOI: 10.3390/cells10040932] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Bone is a hard-vascularized tissue, which renews itself continuously to adapt to the mechanical and metabolic demands of the body. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. The "gold standard" for treating these large defects is autologous bone grafting, which has some drawbacks including the requirement for a second surgical site with quantity of bone limitations, pain and other surgical complications. Indeed, tissue engineering combining a biomaterial with the appropriate cells and molecules of interest would allow a new therapeutic approach to treat large bone defects while avoiding complications associated with a second surgical site. This review first outlines the current knowledge of bone remodeling and the different signaling pathways involved seeking to improve our understanding of the roles of each to be able to stimulate or inhibit them. Secondly, it highlights the interesting characteristics of one growth factor in particular, FGF-2, and its role in bone homeostasis, before then analyzing its potential usefulness in craniofacial bone tissue engineering because of its proliferative, pro-angiogenic and pro-osteogenic effects depending on its spatial-temporal use, dose and mode of administration.
Collapse
Affiliation(s)
- Anita Novais
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Eirini Chatzopoulou
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
- Département de Parodontologie, Université de Paris, UFR Odontologie-Garancière, 75006 Paris, France
| | - Catherine Chaussain
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Caroline Gorin
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| |
Collapse
|
4
|
Kim H, Jeong JH, Fendereski M, Lee HS, Kang DY, Hur SS, Amirian J, Kim Y, Pham NT, Suh N, Hwang NSY, Ryu S, Yoon JK, Hwang Y. Heparin-Mimicking Polymer-Based In Vitro Platform Recapitulates In Vivo Muscle Atrophy Phenotypes. Int J Mol Sci 2021; 22:ijms22052488. [PMID: 33801235 PMCID: PMC7957884 DOI: 10.3390/ijms22052488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
The cell–cell/cell–matrix interactions between myoblasts and their extracellular microenvironment have been shown to play a crucial role in the regulation of in vitro myogenic differentiation and in vivo skeletal muscle regeneration. In this study, by harnessing the heparin-mimicking polymer, poly(sodium-4-styrenesulfonate) (PSS), which has a negatively charged surface, we engineered an in vitro cell culture platform for the purpose of recapitulating in vivo muscle atrophy-like phenotypes. Our initial findings showed that heparin-mimicking moieties inhibited the fusion of mononucleated myoblasts into multinucleated myotubes, as indicated by the decreased gene and protein expression levels of myogenic factors, myotube fusion-related markers, and focal adhesion kinase (FAK). We further elucidated the underlying molecular mechanism via transcriptome analyses, observing that the insulin/PI3K/mTOR and Wnt signaling pathways were significantly downregulated by heparin-mimicking moieties through the inhibition of FAK/Cav3. Taken together, the easy-to-adapt heparin-mimicking polymer-based in vitro cell culture platform could be an attractive platform for potential applications in drug screening, providing clear readouts of changes in insulin/PI3K/mTOR and Wnt signaling pathways.
Collapse
Affiliation(s)
- Hyunbum Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Mona Fendereski
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Hyo-Shin Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Da Yeon Kang
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
| | - Jhaleh Amirian
- Institute of Tissue Regeneration, Soonchunhyang University, Asan-si 31538, Korea;
| | - Yunhye Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nghia Thi Pham
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nayoung Suh
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Nathaniel Suk-Yeon Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| |
Collapse
|
5
|
Im JH, Buzzelli JN, Jones K, Franchini F, Gordon-Weeks A, Markelc B, Chen J, Kim J, Cao Y, Muschel RJ. FGF2 alters macrophage polarization, tumour immunity and growth and can be targeted during radiotherapy. Nat Commun 2020; 11:4064. [PMID: 32792542 PMCID: PMC7426415 DOI: 10.1038/s41467-020-17914-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Regulation of the programming of tumour-associated macrophages (TAMs) controls tumour growth and anti-tumour immunity. We examined the role of FGF2 in that regulation. Tumours in mice genetically deficient in low-molecular weight FGF2 (FGF2LMW) regress dependent on T cells. Yet, TAMS not T cells express FGF receptors. Bone marrow derived-macrophages from Fgf2LMW−/− mice co-injected with cancer cells reduce tumour growth and express more inflammatory cytokines. FGF2 is induced in the tumour microenvironment following fractionated radiation in murine tumours consistent with clinical reports. Combination treatment of in vivo tumours with fractionated radiation and a blocking antibody to FGF2 prolongs tumour growth delay, increases long-term survival and leads to a higher iNOS+/CD206+ TAM ratio compared to irradiation alone. These studies show for the first time that FGF2 affects macrophage programming and is a critical regulator of immunity in the tumour microenvironment. Macrophages contribute to tumour progression and response to therapy. Here, the authors show that absence of FGF2 in the tumour microenvironment reduces tumour growth and enhances the anti-tumour immune response by altering macrophage polarization. As a result, disruption of this macrophage programming by anti-FGF2 blocking antibodies enhances the outcome from radiotherapy.
Collapse
Affiliation(s)
- Jae Hong Im
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jon N Buzzelli
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Keaton Jones
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Fanny Franchini
- The Kennedy Institute of Rheumatology, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Bostjan Markelc
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jianzhou Chen
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jin Kim
- Galaxy Biotech, 1230 Bordeaux Dr, Sunnyvale, CA, 94089, USA
| | - Yunhong Cao
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Ruth J Muschel
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
6
|
Receptor tyrosine kinases and heparan sulfate proteoglycans: Interplay providing anticancer targeting strategies and new therapeutic opportunities. Biochem Pharmacol 2020; 178:114084. [DOI: 10.1016/j.bcp.2020.114084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
|
7
|
Benington L, Rajan G, Locher C, Lim LY. Fibroblast Growth Factor 2-A Review of Stabilisation Approaches for Clinical Applications. Pharmaceutics 2020; 12:E508. [PMID: 32498439 PMCID: PMC7356611 DOI: 10.3390/pharmaceutics12060508] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
Basic fibroblast growth factor (FGF)-2 has been shown to regulate many cellular functions including cell proliferation, migration, and differentiation, as well as angiogenesis in a variety of tissues, including skin, blood vessel, muscle, adipose, tendon/ligament, cartilage, bone, tooth, and nerve. These multiple functions make FGF-2 an attractive component for wound healing and tissue engineering constructs; however, the stability of FGF-2 is widely accepted to be a major concern for the development of useful medicinal products. Many approaches have been reported in the literature for preserving the biological activity of FGF-2 in aqueous solutions. Most of these efforts were directed at sustaining FGF-2 activity for cell culture research, with a smaller number of studies seeking to develop sustained release formulations of FGF-2 for tissue engineering applications. The stabilisation approaches may be classified into the broad classes of ionic interaction modification with excipients, chemical modification, and physical adsorption and encapsulation with carrier materials. This review discusses the underlying causes of FGF-2 instability and provides an overview of the approaches reported in the literature for stabilising FGF-2 that may be relevant for clinical applications. Although efforts have been made to stabilise FGF-2 for both in vitro and in vivo applications with varying degrees of success, the lack of comprehensive published stability data for the final FGF-2 products represents a substantial gap in the current knowledge, which has to be addressed before viable products for wider tissue engineering applications can be developed to meet regulatory authorisation.
Collapse
Affiliation(s)
- Leah Benington
- Division of Pharmacy, School of Allied Health, University of Western Australia, Crawley 6009, Australia; (L.B.); (C.L.)
| | - Gunesh Rajan
- Division of Surgery, School of Medicine, University of Western Australia, Crawley 6009, Australia;
- Department of Otolaryngology, Head & Neck Surgery, Luzerner Kantonsspital, 6000 Luzern, Switzerland
| | - Cornelia Locher
- Division of Pharmacy, School of Allied Health, University of Western Australia, Crawley 6009, Australia; (L.B.); (C.L.)
| | - Lee Yong Lim
- Division of Pharmacy, School of Allied Health, University of Western Australia, Crawley 6009, Australia; (L.B.); (C.L.)
| |
Collapse
|
8
|
Neves MI, Araújo M, Moroni L, da Silva RM, Barrias CC. Glycosaminoglycan-Inspired Biomaterials for the Development of Bioactive Hydrogel Networks. Molecules 2020; 25:E978. [PMID: 32098281 PMCID: PMC7070556 DOI: 10.3390/molecules25040978] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023] Open
Abstract
Glycosaminoglycans (GAG) are long, linear polysaccharides that display a wide range of relevant biological roles. Particularly, in the extracellular matrix (ECM) GAG specifically interact with other biological molecules, such as growth factors, protecting them from proteolysis or inhibiting factors. Additionally, ECM GAG are partially responsible for the mechanical stability of tissues due to their capacity to retain high amounts of water, enabling hydration of the ECM and rendering it resistant to compressive forces. In this review, the use of GAG for developing hydrogel networks with improved biological activity and/or mechanical properties is discussed. Greater focus is given to strategies involving the production of hydrogels that are composed of GAG alone or in combination with other materials. Additionally, approaches used to introduce GAG-inspired features in biomaterials of different sources will also be presented.
Collapse
Affiliation(s)
- Mariana I. Neves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.I.N.); (M.A.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia da Universidade do Porto, Departamento de Engenharia Metalúrgica e de Materiais, Rua Dr Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Marco Araújo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.I.N.); (M.A.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands;
| | - Ricardo M.P. da Silva
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.I.N.); (M.A.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Cristina C. Barrias
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.I.N.); (M.A.)
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
9
|
Bray C, Gurnani P, Mansfield EDH, Peltier R, Perrier S. Sulfonated Copolymers as Heparin-Mimicking Stabilizer of Fibroblast Growth Factor: Size, Architecture, and Monomer Distribution Effects. Biomacromolecules 2019; 20:285-293. [PMID: 30543415 DOI: 10.1021/acs.biomac.8b01451] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fibroblast growth factors (FGF) are involved in a wide range of biological processes such as cell proliferation and differentiation. In living organisms, the binding of FGF to its receptors are mediated through electrostatic interactions between FGF and naturally occurring heparin. Despite its prevalent use in medicine, heparin carries notable limitations; namely, its extraction from natural sources (expensive, low yield and extensive purification), viral contamination, and batch-to-batch heterogeneity. In this work a range of synthetic homopolymers and copolymers of sodium 2-acrylamido-2-methylpropanesulfonate were evaluated as potential FGF stabilizers. This was studied by measuring the proliferation of BaF3-FR1c cells, as a model assay, and the results will be compared with the natural stabilization and activation of FGF by heparin. This study explores the structure-activity relationship of these polysulfonated polymers with a focus on the effect of molecular weight, comonomer type, charge dispersion, and polymer architecture on protein stabilization.
Collapse
Affiliation(s)
- Caroline Bray
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , United Kingdom
| | - Pratik Gurnani
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , United Kingdom
| | - Edward D H Mansfield
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , United Kingdom
| | - Raoul Peltier
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , United Kingdom
| | - Sébastien Perrier
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , United Kingdom
- Warwick Medical School , University of Warwick , Gibbet Hill Road , Coventry CV4 7AL , United Kingdom
- Faculty of Pharmacy and Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| |
Collapse
|
10
|
Jiang S, Wu J, Hang Y, Liu Q, Li D, Chen H, Brash JL. Sustained release of a synthetic structurally-tailored glycopolymer modulates endothelial cells for enhanced endothelialization of materials. J Mater Chem B 2019. [DOI: 10.1039/c9tb00714h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
GAG-mimicking polymers were prepared by a novel method allowing close control of structure and can be used as potent synthetic bioactive modifiers to promote endothelialization of materials.
Collapse
Affiliation(s)
- Shuaibing Jiang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Jingxian Wu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Yingjie Hang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Qi Liu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Dan Li
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - John L. Brash
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| |
Collapse
|
11
|
Abstract
This review describes several general chemical approaches for the preparation of glycosaminoglycan (GAG)-mimetic polymers based on different backbones and sidechains, and highlights the importance of these synthetic GAG-mimetic polymers in controlling key biofunctions.
Collapse
Affiliation(s)
- Qi Liu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Gaojian Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
| |
Collapse
|
12
|
Tomic-Canic M, Wong LL, Smola H. The epithelialisation phase in wound healing: options to enhance wound closure. J Wound Care 2018; 27:646-658. [DOI: 10.12968/jowc.2018.27.10.646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Majana Tomic-Canic
- Professor and Vice Chair of Research; Director, Wound Healing and Regenerative Medicine Research Program; Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, Florida, US
| | - Lulu L. Wong
- MD Candidate; Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, Florida, US
| | - Hans Smola
- Professor of Dermatology, Medical Director, PAUL HARTMANN AG, Heidenheim and Department of Dermatology, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Lei J, Yuan Y, Lyu Z, Wang M, Liu Q, Wang H, Yuan L, Chen H. Deciphering the Role of Sulfonated Unit in Heparin-Mimicking Polymer to Promote Neural Differentiation of Embryonic Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:28209-28221. [PMID: 28783314 DOI: 10.1021/acsami.7b08034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Glycosaminoglycans (GAGs), especially heparin and heparan sulfate (HS), hold great potential for inducing the neural differentiation of embryonic stem cells (ESCs) and have brought new hope for the treatment of neurological diseases. However, the disadvantages of natural heparin/HS, such as difficulty in isolating them with a sufficient amount, highly heterogeneous structure, and the risk of immune responses, have limited their further therapeutic applications. Thus, there is a great demand for stable, controllable, and well-defined synthetic alternatives of heparin/HS with more effective biological functions. In this study, based upon a previously proposed unit-recombination strategy, several heparin-mimicking polymers were synthesized by integrating glucosamine-like 2-methacrylamido glucopyranose monomers (MAG) with three sulfonated units in different structural forms, and their effects on cell proliferation, the pluripotency, and the differentiation of ESCs were carefully studied. The results showed that all the copolymers had good cytocompatibility and displayed much better bioactivity in promoting the neural differentiation of ESCs as compared to natural heparin; copolymers with different sulfonated units exhibited different levels of promoting ability; among them, copolymer with 3-sulfopropyl acrylate (SPA) as a sulfonated unit was the most potent in promoting the neural differentiation of ESCs; the promoting effect is dependent on the molecular weight and concentration of P(MAG-co-SPA), with the highest levels occurring at the intermediate molecular weight and concentration. These results clearly demonstrated that the sulfonated unit in the copolymers played an important role in determining the promoting effect on ESCs' neural differentiation; SPA was identified as the most potent sulfonated unit for copolymer with the strongest promoting ability. The possible reason for sulfonated unit structure as a vital factor influencing the ability of the copolymers may be attributed to the difference in electrostatic and steric hindrance effect. The synthetic heparin-mimicking polymers obtained here can offer an effective alternative to heparin/HS and have great therapeutic potential for nervous system diseases.
Collapse
Affiliation(s)
- Jiehua Lei
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Yuqi Yuan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Zhonglin Lyu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Mengmeng Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Qi Liu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Hongwei Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Lin Yuan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| |
Collapse
|
14
|
Paluck SJ, Maynard HD. Structure Activity Relationship of Heparin Mimicking Polymer p(SS- co-PEGMA): Effect of Sulfonation and Polymer Size on FGF2-Receptor Binding. Polym Chem 2017; 8:4548-4556. [PMID: 29181096 PMCID: PMC5701782 DOI: 10.1039/c7py00861a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fibroblast growth factor-2 (FGF2) is a heparin binding protein that plays a role in a range of biological functions such as wound healing and bone regeneration. Heparin, a highly sulfated glycosaminoglycan, is required for FGF2 to bind to its receptor. Therefore, polymeric mimics of heparin are widely studied for their ability to manipulate FGF2-induced biological interactions. It is known that altering the degree of sulfonated monomer incorporation and size of heparin-mimicking polymers can affect protein-receptor binding. To elucidate the relationship between degree of sulfonation and receptor binding for the heparin-mimicking polymer, poly(styrene sulfonate-co-poly(ethylene glycol) methyl ether methacrylate) (p(SS-co-PEGMA)) a library was synthesized to contain nine polymers with degrees of sulfonation ranging from 0-100%. Kinetics of the polymerization was evaluated and reactivity ratios compared to literature results. These polymers were then tested for their ability to enhance FGF2 binding with its receptor as both covalent conjugates and as excipients. In a receptor based enzyme-linked immunosorbant assay (ELISA), as well as a cell-based study, the polymer with 81% SS incorporation enhanced receptor binding compared to FGF2 alone, and to a greater extent than the other polymers. Therefore, another library of polymers was prepared maintaining the degree of sulfonation at 81% and changing the size from 41 to 390 monomer repeat units. The polymers were again tested in receptor based ELISA and cell studies, and all of the different sizes performed similarly, except for degree of polymerization 295 and 390, which had reduced response in the cellular assay. These results provide important information for the use of pSS-co-PEGMA as a potential heparin-mimicking therapeutic.
Collapse
Affiliation(s)
- Samantha J Paluck
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Heather D Maynard
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| |
Collapse
|
15
|
Paluck S, Nguyen TH, Maynard HD. Heparin-Mimicking Polymers: Synthesis and Biological Applications. Biomacromolecules 2016; 17:3417-3440. [PMID: 27739666 PMCID: PMC5111123 DOI: 10.1021/acs.biomac.6b01147] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/26/2016] [Indexed: 12/13/2022]
Abstract
Heparin is a naturally occurring, highly sulfated polysaccharide that plays a critical role in a range of different biological processes. Therapeutically, it is mostly commonly used as an injectable solution as an anticoagulant for a variety of indications, although it has also been employed in other forms such as coatings on various biomedical devices. Due to the diverse functions of this polysaccharide in the body, including anticoagulation, tissue regeneration, anti-inflammation, and protein stabilization, and drawbacks of its use, analogous heparin-mimicking materials are also widely studied for therapeutic applications. This review focuses on one type of these materials, namely, synthetic heparin-mimicking polymers. Utilization of these polymers provides significant benefits compared to heparin, including enhancing therapeutic efficacy and reducing side effects as a result of fine-tuning heparin-binding motifs and other molecular characteristics. The major types of the various polymers are summarized, as well as their applications. Because development of a broader range of heparin-mimicking materials would further expand the impact of these polymers in the treatment of various diseases, future directions are also discussed.
Collapse
Affiliation(s)
- Samantha
J. Paluck
- Department of Chemistry and
Biochemistry and the California NanoSystems Institute, University of California−Los Angeles, 607 Charles E. Young Dr East, Los Angeles, California 90095, United States
| | - Thi H. Nguyen
- Department of Chemistry and
Biochemistry and the California NanoSystems Institute, University of California−Los Angeles, 607 Charles E. Young Dr East, Los Angeles, California 90095, United States
| | - Heather D. Maynard
- Department of Chemistry and
Biochemistry and the California NanoSystems Institute, University of California−Los Angeles, 607 Charles E. Young Dr East, Los Angeles, California 90095, United States
| |
Collapse
|
16
|
Paluck S, Nguyen TH, Lee JP, Maynard HD. A Heparin-Mimicking Block Copolymer Both Stabilizes and Increases the Activity of Fibroblast Growth Factor 2 (FGF2). Biomacromolecules 2016; 17:3386-3395. [PMID: 27580376 PMCID: PMC5059753 DOI: 10.1021/acs.biomac.6b01182] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/27/2016] [Indexed: 01/22/2023]
Abstract
Fibroblast growth factor 2 (FGF2) is a protein involved in cellular functions in applications such as wound healing and tissue regeneration. Stabilization of this protein is important for its use as a therapeutic since the native protein is unstable during storage and delivery. Additionally, the ability to increase the activity of FGF2 is important for its application, particularly in chronic wound healing and the treatment of various ischemic conditions. Here we report a heparin mimicking block copolymer, poly(styrenesulfonate-co-poly(ethylene glycol) methyl ether methacrylate)-b-vinyl sulfonate) (p(SS-co-PEGMA)-b-VS, that contains a segment that enhances the stability of FGF2 and one that binds to the FGF2 receptor. The FGF2 conjugate retained activity after exposure to refrigeration (4 °C) and room temperature (23 °C) for 7 days, while unmodified FGF2 was inactive after these standard storage conditions. A cell study performed with a cell line lacking native heparan sulfate proteoglycans indicated that the conjugated block copolymer facilitated binding of FGF2 to its receptor similar to the addition of heparin to FGF2. A receptor-based enzyme-linked immunosorbant assay (ELISA) confirmed the results. The conjugate also increased the migration of endothelial cells by 80% compared to FGF2 alone. Additionally, the FGF2-p(SS-co-PEGMA)-b-VS stimulated endothelial cell sprouting 250% better than FGF2 at low concentration. These data verify that this rationally designed protein-block copolymer conjugate enhances receptor binding, cellular processes such as migration and tube-like formation, and stability, and suggest that it may be useful for applications in biomaterials, tissue regeneration, and wound healing.
Collapse
Affiliation(s)
- Samantha
J. Paluck
- Department of Chemistry and
Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Thi H. Nguyen
- Department of Chemistry and
Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Jonghan P. Lee
- Department of Chemistry and
Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Heather D. Maynard
- Department of Chemistry and
Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| |
Collapse
|
17
|
Avizienyte E, Cole CL, Rushton G, Miller GJ, Bugatti A, Presta M, Gardiner JM, Jayson GC. Synthetic Site-Selectively Mono-6-O-Sulfated Heparan Sulfate Dodecasaccharide Shows Anti-Angiogenic Properties In Vitro and Sensitizes Tumors to Cisplatin In Vivo. PLoS One 2016; 11:e0159739. [PMID: 27490176 PMCID: PMC4973927 DOI: 10.1371/journal.pone.0159739] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/07/2016] [Indexed: 11/23/2022] Open
Abstract
Heparan sulphate (HS), a ubiquitously expressed glycosaminoglycan (GAG), regulates multiple cellular functions by mediating interactions between numerous growth factors and their cell surface cognate receptors. However, the structural specificity of HS in these interactions remains largely undefined. Here, we used completely synthetic, structurally defined, alternating N-sulfated glucosamine (NS) and 2-O-sulfated iduronate (IS) residues to generate dodecasaccharides ([NSIS]6) that contained no, one or six glucosamine 6-O-sulfates (6S). The aim was to address how 6S contributes to the potential of defined HS dodecasaccharides to inhibit the angiogenic growth factors FGF2 and VEGF165, in vitro and in vivo. We show that the addition of a single 6S at the non-reducing end of [NSIS]6, i.e. [NSIS6S]-[NSIS]5, significantly augments the inhibition of FGF2-dependent endothelial cell proliferation, migration and sprouting in vitro when compared to the non-6S variant. In contrast, the fully 6-O-sulfated dodecasaccharide, [NSIS6S]6, is not a potent inhibitor of FGF2. Addition of a single 6S did not significantly improve inhibitory properties of [NSIS]6 when tested against VEGF165-dependent endothelial cell functions.In vivo, [NSIS6S]-[NSIS]5 blocked FGF2-dependent blood vessel formation without affecting tumor growth. Reduction of non-FGF2-dependent ovarian tumor growth occurred when [NSIS6S]-[NSIS]5 was combined with cisplatin. The degree of inhibition by [NSIS6S]-[NSIS]5 in combination with cisplatin in vivo equated with that induced by bevacizumab and sunitinib when administered with cisplatin. Evaluation of post-treatment vasculature revealed that [NSIS6S]-[NSIS]5 treatment had the greatest impact on tumor blood vessel size and lumen formation. Our data for the first time demonstrate that synthetic, structurally defined oligosaccharides have potential to be developed as active anti-angiogenic agents that sensitize tumors to chemotherapeutic agents.
Collapse
Affiliation(s)
- Egle Avizienyte
- Institute of Cancer Sciences, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
- * E-mail: (EA); (GCJ)
| | - Claire L. Cole
- Institute of Cancer Sciences, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
| | - Graham Rushton
- Institute of Cancer Sciences, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
| | - Gavin J. Miller
- School of Chemistry and Manchester Interdisciplinary Biocentre, The University of Manchester, Manchester M1 7ND, United Kingdom
| | - Antonella Bugatti
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - John M. Gardiner
- School of Chemistry and Manchester Interdisciplinary Biocentre, The University of Manchester, Manchester M1 7ND, United Kingdom
| | - Gordon C. Jayson
- Institute of Cancer Sciences, Faculty of Medical and Human Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
- * E-mail: (EA); (GCJ)
| |
Collapse
|
18
|
Akl MR, Nagpal P, Ayoub NM, Tai B, Prabhu SA, Capac CM, Gliksman M, Goy A, Suh KS. Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget 2016; 7:44735-44762. [PMID: 27007053 PMCID: PMC5190132 DOI: 10.18632/oncotarget.8203] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/10/2016] [Indexed: 12/30/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling is essential for normal and cancer biology. Mammalian FGF family members participate in multiple signaling pathways by binding to heparan sulfate and FGF receptors (FGFR) with varying affinities. FGF2 is the prototype member of the FGF family and interacts with its receptor to mediate receptor dimerization, phosphorylation, and activation of signaling pathways, such as Ras-MAPK and PI3K pathways. Excessive mitogenic signaling through the FGF/FGFR axis may induce carcinogenic effects by promoting cancer progression and increasing the angiogenic potential, which can lead to metastatic tumor phenotypes. Dysregulated FGF/FGFR signaling is associated with aggressive cancer phenotypes, enhanced chemotherapy resistance and poor clinical outcomes. In vitro experimental settings have indicated that extracellular FGF2 affects proliferation, drug sensitivity, and apoptosis of cancer cells. Therapeutically targeting FGF2 and FGFR has been extensively assessed in multiple preclinical studies and numerous drugs and treatment options have been tested in clinical trials. Diagnostic assays are used to quantify FGF2, FGFRs, and downstream signaling molecules to better select a target patient population for higher efficacy of cancer therapies. This review focuses on the prognostic significance of FGF2 in cancer with emphasis on therapeutic intervention strategies for solid and hematological malignancies.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Catherine M. Capac
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
19
|
Öztürk E, Arlov Ø, Aksel S, Li L, Ornitz DM, Skjåk-Bræk G, Zenobi-Wong M. Sulfated hydrogel matrices direct mitogenicity and maintenance of chondrocyte phenotype through activation of FGF signaling. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3649-3662. [PMID: 28919847 PMCID: PMC5597002 DOI: 10.1002/adfm.201600092] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Deciphering the roles of chemical and physical features of the extracellular matrix (ECM) is vital for developing biomimetic materials with desired cellular responses in regenerative medicine. Here, we demonstrate that sulfation of biopolymers, mimicking the proteoglycans in native tissues, induces mitogenicity, chondrogenic phenotype, and suppresses catabolic activity of chondrocytes, a cell type that resides in a highly sulfated tissue. We show through tunable modification of alginate that increased sulfation of the microenvironment promotes FGF signaling-mediated proliferation of chondrocytes in a three-dimensional (3D) matrix independent of stiffness, swelling, and porosity. Furthermore, we show for the first time that a biomimetic hydrogel acts as a 3D signaling matrix to mediate a heparan sulfate/heparin-like interaction between FGF and its receptor leading to signaling cascades inducing cell proliferation, cartilage matrix production, and suppression of de-differentiation markers. Collectively, this study reveals important insights on mimicking the ECM to guide self-renewal of cells via manipulation of distinct signaling mechanisms.
Collapse
Affiliation(s)
- Ece Öztürk
- Cartilage Engineering+ Regeneration, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Øystein Arlov
- Department of Biotechnology, Norwegian University of Science and Technology, Sem Sælands vei 6/8, 7034 Trondheim, Norway
| | - Seda Aksel
- Department of Materials, Polymer Technology Laboratory, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Ling Li
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gudmund Skjåk-Bræk
- Department of Biotechnology, Norwegian University of Science and Technology, Sem Sælands vei 6/8, 7034 Trondheim, Norway
| | | |
Collapse
|
20
|
Integrating computational and chemical biology tools in the discovery of antiangiogenic small molecule ligands of FGF2 derived from endogenous inhibitors. Sci Rep 2016; 6:23432. [PMID: 27000667 PMCID: PMC4802308 DOI: 10.1038/srep23432] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/07/2016] [Indexed: 01/22/2023] Open
Abstract
The FGFs/FGFRs system is a recognized actionable target for therapeutic approaches aimed at inhibiting tumor growth, angiogenesis, metastasis, and resistance to therapy. We previously identified a non-peptidic compound (SM27) that retains the structural and functional properties of the FGF2-binding sequence of thrombospondin-1 (TSP-1), a major endogenous inhibitor of angiogenesis. Here we identified new small molecule inhibitors of FGF2 based on the initial lead. A similarity-based screening of small molecule libraries, followed by docking calculations and experimental studies, allowed selecting 7 bi-naphthalenic compounds that bound FGF2 inhibiting its binding to both heparan sulfate proteoglycans and FGFR-1. The compounds inhibit FGF2 activity in in vitro and ex vivo models of angiogenesis, with improved potency over SM27. Comparative analysis of the selected hits, complemented by NMR and biochemical analysis of 4 newly synthesized functionalized phenylamino-substituted naphthalenes, allowed identifying the minimal stereochemical requirements to improve the design of naphthalene sulfonates as FGF2 inhibitors.
Collapse
|
21
|
Nie C, Cheng C, Peng Z, Ma L, He C, Xia Y, Zhao C. Mussel-inspired coatings on Ag nanoparticle-conjugated carbon nanotubes: bactericidal activity and mammal cell toxicity. J Mater Chem B 2016; 4:2749-2756. [DOI: 10.1039/c6tb00470a] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Silver nanoparticle (AgNP)-based nanohybrids have been proposed as efficient antimicrobial agents because of their robust bactericidal activity.
Collapse
Affiliation(s)
- Chuanxiong Nie
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| | - Chong Cheng
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| | - Zihang Peng
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| | - Lang Ma
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| | - Chao He
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| | - Yi Xia
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| | - Changsheng Zhao
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu
- China
| |
Collapse
|
22
|
Nguyen TH, Paluck SJ, McGahran AJ, Maynard HD. Poly(vinyl sulfonate) Facilitates bFGF-Induced Cell Proliferation. Biomacromolecules 2015. [PMID: 26212474 DOI: 10.1021/acs.biomac.5b00557] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Heparin is a highly sulfated polysaccharide and is useful because of its diverse biological functions. However, because of batch-to-batch variability and other factors, there is significant interest in preparing biomimetics of heparin. To identify polymeric heparin mimetics, a cell-based screening assay was developed in cells that express fibroblast growth factor receptors (FGFRs) but not heparan sulfate proteoglycans. Various sulfated and sulfonated polymers were screened, and poly(vinyl sulfonate) (pVS) was identified as the strongest heparin-mimicking polymer in its ability to enhance binding of basic fibroblast growth factor (bFGF) to FGFR. The results were confirmed by an ELISA-based receptor-binding assay. Different molecular weights of pVS polymer were synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization. The polymers were able to facilitate dimerization of FGFRs leading to cell proliferation in FGFR-expressing cells, and no size dependence was observed. The data showed that pVS is comparable to heparin in these assays. In addition, pVS was not cytotoxic to fibroblast cells up to at least 1 mg/mL. Together this data indicates that pVS should be explored further as a replacement for heparin.
Collapse
Affiliation(s)
- Thi H Nguyen
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles , 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Samantha J Paluck
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles , 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Andrew J McGahran
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles , 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| | - Heather D Maynard
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles , 607 Charles E. Young Drive East, Los Angeles, California 90095-1569 United States
| |
Collapse
|
23
|
Niu TT, Zhang DS, Chen HM, Yan XJ. Modulation of the binding of basic fibroblast growth factor and heparanase activity by purified λ-carrageenan oligosaccharides. Carbohydr Polym 2015; 125:76-84. [DOI: 10.1016/j.carbpol.2015.02.069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/06/2015] [Accepted: 02/28/2015] [Indexed: 12/28/2022]
|
24
|
Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: biological implications and therapeutical use. Molecules 2015; 20:6342-88. [PMID: 25867824 PMCID: PMC6272510 DOI: 10.3390/molecules20046342] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis, the process of formation of new blood vessel from pre-existing ones, is involved in various intertwined pathological processes including virus infection, inflammation and oncogenesis, making it a promising target for the development of novel strategies for various interventions. To induce angiogenesis, angiogenic growth factors (AGFs) must interact with pro-angiogenic receptors to induce proliferation, protease production and migration of endothelial cells (ECs). The action of AGFs is counteracted by antiangiogenic modulators whose main mechanism of action is to bind (thus sequestering or masking) AGFs or their receptors. Many sugars, either free or associated to proteins, are involved in these interactions, thus exerting a tight regulation of the neovascularization process. Heparin and heparan sulfate proteoglycans undoubtedly play a pivotal role in this context since they bind to almost all the known AGFs, to several pro-angiogenic receptors and even to angiogenic inhibitors, originating an intricate network of interaction, the so called "angiogenesis glycomic interactome". The decoding of the angiogenesis glycomic interactome, achievable by a systematic study of the interactions occurring among angiogenic modulators and sugars, may help to design novel antiangiogenic therapies with implications in the cure of angiogenesis-dependent diseases.
Collapse
|
25
|
Wang M, Lyu Z, Chen G, Wang H, Yuan Y, Ding K, Yu Q, Yuan L, Chen H. A new avenue to the synthesis of GAG-mimicking polymers highly promoting neural differentiation of embryonic stem cells. Chem Commun (Camb) 2015; 51:15434-7. [DOI: 10.1039/c5cc06944k] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A new strategy for the fabrication of glycosaminoglycan (GAG) analogs with high bioactivities was proposed by copolymerizing the sulfonated unit and the glyco unit, ‘splitted’ from the sulfated saccharide building blocks of GAGs.
Collapse
Affiliation(s)
- Mengmeng Wang
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Zhonglin Lyu
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Gaojian Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research
- Soochow University
- Suzhou 215006
- China
| | - Hongwei Wang
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Yuqi Yuan
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Kaiguo Ding
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Qian Yu
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Lin Yuan
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| | - Hong Chen
- College of Chemistry
- Chemical Engineering and Materials Science
- Soochow University
- Suzhou 215123
- China
| |
Collapse
|
26
|
Kim HS, Yoo HS. Surface-polymerized biomimetic nanofibrils for the cell-directed association of 3-D scaffolds. Chem Commun (Camb) 2014; 51:306-9. [PMID: 25406987 DOI: 10.1039/c4cc06875k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
For the cell-directed association of nanofibrils, polymeric nanofibrils (NFs) were fabricated by fragmentation of electrospun nanofibers and subsequent surface-decoration with poly(methacrylate) derivatives via ATRP. The decorated NFs showed higher degrees of protein sequestration and enhanced cell proliferation with free infiltration of cells, which leads to hydrogel-like behavior.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Biomedical Materials Engineering, College of Biomedical Science, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| | | |
Collapse
|
27
|
Peña B, Shandas R, Park D. A heparin-mimicking reverse thermal gel for controlled delivery of positively charged proteins. J Biomed Mater Res A 2014; 103:2102-8. [PMID: 25294242 DOI: 10.1002/jbm.a.35345] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/23/2014] [Accepted: 09/30/2014] [Indexed: 12/13/2022]
Abstract
Positively charged therapeutic proteins have been used extensively for biomedical applications. However, the safety and efficacy of proteins are mostly limited by their physical and chemical instability and short half-lives in physiological conditions. To this end, we created a heparin-mimicking sulfonated reverse thermal gel as a novel protein delivery system by sulfonation of a graft copolymer, poly(serinol hexamethylene urea)-co-poly(N-isopropylacylamide), or PSHU-NIPAAm. The net charge of the sulfonated PSHU-NIPAAm was negative due to the presence of sulfonate groups. The sulfonated PSHU-NIPAAm showed a typical temperature-dependent sol-gel phase transition, where polymer solutions turned to a physical gel at around 32°C and maintained gel status at body temperature. Both in vitro cytotoxicity tests using C2C12 myoblast cells and in vivo cytotoxicity tests by subcutaneous injections demonstrated excellent biocompatibility. In vitro release tests using bovine serum albumin revealed that the release from the sulfonated PSHU-NIPAAm was more sustained than that from the plain PSHU-NIPAAm. Furthermore, this sulfonated PSHU-NIPAAm system did not affect protein structure after 70-day observation periods.
Collapse
Affiliation(s)
- Brisa Peña
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, 80045
| | | | | |
Collapse
|
28
|
Nguyen TH, Kim SH, Decker CG, Wong DY, Loo JA, Maynard HD. A heparin-mimicking polymer conjugate stabilizes basic fibroblast growth factor. Nat Chem 2013; 5:221-7. [PMID: 23422564 PMCID: PMC3579505 DOI: 10.1038/nchem.1573] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/14/2013] [Indexed: 12/18/2022]
Abstract
Basic fibroblast growth factor (bFGF) is a protein that plays a crucial role in diverse cellular functions, from wound healing to bone regeneration. However, a major obstacle to the widespread application of bFGF is its inherent instability during storage and delivery. Here, we describe the stabilization of bFGF by covalent conjugation with a heparin-mimicking polymer, a copolymer consisting of styrene sulfonate units and methyl methacrylate units bearing poly(ethylene glycol) side chains. The bFGF conjugate of this polymer retained bioactivity after synthesis and was stable to a variety of environmentally and therapeutically relevant stressors--such as heat, mild and harsh acidic conditions, storage and proteolytic degradation--unlike native bFGF. Following the application of stress, the conjugate was also significantly more active than the control conjugate system in which the styrene sulfonate units were omitted from the polymer structure. This research has important implications for the clinical use of bFGF and for the stabilization of heparin-binding growth factors in general.
Collapse
Affiliation(s)
- Thi H. Nguyen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Sung-Hye Kim
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Caitlin G. Decker
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Darice Y. Wong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Tardajos MG, García-Fernández L, Reinecke H, Aguilar MR, Gallardo A, Román JS. Microstructure and biological activity of sulfonated N-vinylpyrrolidone copolymers. J BIOACT COMPAT POL 2012. [DOI: 10.1177/0883911512457761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article deals with the preparation, characterization, and in vitro evaluation of new copolymers based on functionalized N-vinylpyrrolidone bearing pendant sulfonic acid groups statistically or alternately distributed in the macromolecular chains. Statistical copolymers were obtained by free radical polymerization of N-vinylpyrrolidone and sulfonated N-vinylpyrrolidone ( r VP = 1.61; r VPSulf = 0.90) in water, and alternating copolymers were synthesized by free radical polymerization of N-vinylpyrrolidone or sulfonated N-vinylpyrrolidone with methyl maleate in N,N-dimethylformamide followed by hydrolysis of the resulting copolymers. The influence of these new materials on acidic fibroblast growth factor–mediated mitogenesis of fibroblasts demonstrated that sequence distribution (copolymer microstructure) plays a key role in the biological performance of these polymers. Alternating copolymers did not present biological activity, whereas statistical copolymers inhibited acidic fibroblast growth factor activity in a sulfonated N-vinylpyrrolidone dose-dependent manner.
Collapse
Affiliation(s)
- Myriam G Tardajos
- Polymer Functionalization Group, Polymer Physics, Elastomers and Energy Department, Polymer Science and Technology Institute (ICTP-CSIC), Madrid, Spain
| | - Luis García-Fernández
- Biomaterials Group, Polymeric Nanomaterials and Biomaterials Department, Polymer Science and Technology Institute (ICTP-CSIC), Madrid, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - Helmut Reinecke
- Polymer Functionalization Group, Polymer Physics, Elastomers and Energy Department, Polymer Science and Technology Institute (ICTP-CSIC), Madrid, Spain
| | - María R Aguilar
- Biomaterials Group, Polymeric Nanomaterials and Biomaterials Department, Polymer Science and Technology Institute (ICTP-CSIC), Madrid, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - Alberto Gallardo
- Polymer Functionalization Group, Polymer Physics, Elastomers and Energy Department, Polymer Science and Technology Institute (ICTP-CSIC), Madrid, Spain
| | - Julio S Román
- Biomaterials Group, Polymeric Nanomaterials and Biomaterials Department, Polymer Science and Technology Institute (ICTP-CSIC), Madrid, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
30
|
Liu L, Li C, Cochran S, Jimmink S, Ferro V. Synthesis of a Heparan Sulfate Mimetic Library Targeting FGF and VEGF via Click Chemistry on a Monosaccharide Template. ChemMedChem 2012; 7:1267-75. [DOI: 10.1002/cmdc.201200151] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 04/26/2012] [Indexed: 11/07/2022]
|
31
|
Pagano K, Torella R, Foglieni C, Bugatti A, Tomaselli S, Zetta L, Presta M, Rusnati M, Taraboletti G, Colombo G, Ragona L. Direct and allosteric inhibition of the FGF2/HSPGs/FGFR1 ternary complex formation by an antiangiogenic, thrombospondin-1-mimic small molecule. PLoS One 2012; 7:e36990. [PMID: 22606323 PMCID: PMC3351436 DOI: 10.1371/journal.pone.0036990] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 04/11/2012] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factors (FGFs) are recognized targets for the development of therapies against angiogenesis-driven diseases, including cancer. The formation of a ternary complex with the transmembrane tyrosine kinase receptors (FGFRs), and heparan sulphate proteoglycans (HSPGs) is required for FGF2 pro-angiogenic activity. Here by using a combination of techniques including Nuclear Magnetic Resonance, Molecular Dynamics, Surface Plasmon Resonance and cell-based binding assays we clarify the molecular mechanism of inhibition of an angiostatic small molecule, sm27, mimicking the endogenous inhibitor of angiogenesis, thrombospondin-1. NMR and MD data demonstrate that sm27 engages the heparin-binding site of FGF2 and induces long-range dynamics perturbations along FGF2/FGFR1 interface regions. The functional consequence of the inhibitor binding is an impaired FGF2 interaction with both its receptors, as demonstrated by SPR and cell-based binding assays. We propose that sm27 antiangiogenic activity is based on a twofold-direct and allosteric-mechanism, inhibiting FGF2 binding to both its receptors.
Collapse
Affiliation(s)
- Katiuscia Pagano
- Laboratorio NMR, Istituto per lo Studio delle Macromolecole, Consiglio Nazionale delle Ricerche, Milano, Italy
| | - Rubben Torella
- Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche, Milano, Italy
| | - Chiara Foglieni
- Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | - Antonella Bugatti
- Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | - Simona Tomaselli
- Laboratorio NMR, Istituto per lo Studio delle Macromolecole, Consiglio Nazionale delle Ricerche, Milano, Italy
| | - Lucia Zetta
- Laboratorio NMR, Istituto per lo Studio delle Macromolecole, Consiglio Nazionale delle Ricerche, Milano, Italy
| | - Marco Presta
- Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | - Marco Rusnati
- Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | - Giulia Taraboletti
- Department of Oncology, Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | - Giorgio Colombo
- Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche, Milano, Italy
- * E-mail: (LR); (GC)
| | - Laura Ragona
- Laboratorio NMR, Istituto per lo Studio delle Macromolecole, Consiglio Nazionale delle Ricerche, Milano, Italy
- * E-mail: (LR); (GC)
| |
Collapse
|
32
|
Liekens S, Bronckaers A, Belleri M, Bugatti A, Sienaert R, Ribatti D, Nico B, Gigante A, Casanova E, Opdenakker G, Pérez-Pérez MJ, Balzarini J, Presta M. The thymidine phosphorylase inhibitor 5'-O-tritylinosine (KIN59) is an antiangiogenic multitarget fibroblast growth factor-2 antagonist. Mol Cancer Ther 2012; 11:817-29. [PMID: 22302099 DOI: 10.1158/1535-7163.mct-11-0738] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
5'-O-Tritylinosine (KIN59) is an allosteric inhibitor of the angiogenic enzyme thymidine phosphorylase. Previous observations showed the capacity of KIN59 to abrogate thymidine phosphorylase-induced as well as developmental angiogenesis in the chicken chorioallantoic membrane (CAM) assay. Here, we show that KIN59 also inhibits the angiogenic response triggered by fibroblast growth factor-2 (FGF2) but not by VEGF in the CAM assay. Immunohistochemical and reverse transcriptase PCR analyses revealed that the expression of laminin, the major proteoglycan of the basement membrane of blood vessels, is downregulated by KIN59 administration in control as well as in thymidine phosphorylase- or FGF2-treated CAMs, but not in CAMs treated with VEGF. Also, KIN59 abrogated FGF2-induced endothelial cell proliferation, FGF receptor activation, and Akt signaling in vitro with no effect on VEGF-stimulated biologic responses. Accordingly, KIN59 inhibited the binding of FGF2 to FGF receptor-1 (FGFR1), thus preventing the formation of productive heparan sulphate proteoglycan/FGF2/FGFR1 ternary complexes, without affecting heparin interaction. In keeping with these observations, systemic administration of KIN59 inhibited the growth and neovascularization of subcutaneous tumors induced by FGF2-transformed endothelial cells injected in immunodeficient nude mice. Taken together, the data indicate that the thymidine phosphorylase inhibitor KIN59 is endowed with a significant FGF2 antagonist activity, thus representing a promising lead compound for the design of multitargeted antiangiogenic cancer drugs.
Collapse
Affiliation(s)
- Sandra Liekens
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
García-Fernández L, Aguilar MR, Ochoa-Callejero L, Abradelo C, Martínez A, San Román J. bFGF interaction and in vivo angiogenesis inhibition by self-assembling sulfonic acid-based copolymers. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:129-135. [PMID: 22116660 DOI: 10.1007/s10856-011-4497-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 11/09/2011] [Indexed: 05/31/2023]
Abstract
The antiangiogenic activity of different families of biocompatible and non-toxic polymer drugs based on 2-acrylamido-2-methylpropane sulfonic acid (AMPS) or polymethacrylic derivatives of 5-aminonaphthalen sulfonic acid (MANSA) is analyzed using directed in vivo angiogenesis assay and correlated with in vitro results. These active compounds were copolymerized with butylacrylate (BA) and N-vinylpyrrolidone in order to obtain two families of copolymers with different properties in aqueous media. The most hydrophobic copolymers poly(BA-co-MANSA) and poly(BA-co-AMPS) formed amphiphilic copolymers and presented micellar morphology in aqueous media. This supramolecular organization of the copolymers had a clear effect on bioactivity. Poly(BA-co-MANSA) copolymers showed the best antiangiogenic activity and very low toxicity at relatively low dose, with the possibility to be injected directly in the solid tumors alone or in combination with other therapeutic agents such as anti-VEGF drugs. The obtained results demonstrate that not only the chemical structure but also the supramolecular organization of the macromolecules plays a key role in the anti-angiogenic activity of these active polymers.
Collapse
Affiliation(s)
- L García-Fernández
- Department of Biomaterials, Institute of Polymer Science and Technology, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
34
|
Kolodziej CM, Kim SH, Broyer RM, Saxer SS, Decker CG, Maynard HD. Combination of integrin-binding peptide and growth factor promotes cell adhesion on electron-beam-fabricated patterns. J Am Chem Soc 2011; 134:247-55. [PMID: 22126191 DOI: 10.1021/ja205524x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding and controlling cell adhesion on engineered scaffolds is important in biomaterials and tissue engineering. In this report we used an electron-beam (e-beam) lithography technique to fabricate patterns of a cell adhesive integrin ligand combined with a growth factor. Specifically, micron-sized poly(ethylene glycol) (PEG) hydrogels with aminooxy- and styrene sulfonate-functional groups were fabricated. Cell adhesion moieties were introduced using a ketone-functionalized arginine-glycine-aspartic acid (RGD) peptide to modify the O-hydroxylamines by oxime bond formation. Basic fibroblast growth factor (bFGF) was immobilized by electrostatic interaction with the sulfonate groups. Human umbilical vein endothelial cells (HUVECs) formed focal adhesion complexes on RGD- and RGD and bFGF-immobilized patterns as shown by immunostaining of vinculin and actin. In the presence of both bFGF and RGD, cell areas were larger. The data demonstrate confinement of cellular focal adhesions to chemically and physically well-controlled microenvironments created by a combination of e-beam lithography and "click" chemistry techniques. The results also suggest positive implications for addition of growth factors into adhesive patterns for cell-material interactions.
Collapse
Affiliation(s)
- Christopher M Kolodziej
- Department of Chemistry and Biochemistry and the California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive South, Los Angeles, California 90095, USA
| | | | | | | | | | | |
Collapse
|
35
|
Leali D, Alessi P, Coltrini D, Ronca R, Corsini M, Nardo G, Indraccolo S, Presta M. Long pentraxin-3 inhibits FGF8b-dependent angiogenesis and growth of steroid hormone-regulated tumors. Mol Cancer Ther 2011; 10:1600-10. [PMID: 21764903 DOI: 10.1158/1535-7163.mct-11-0286] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fibroblast growth factor-8b (FGF8b) exerts nonredundant autocrine/paracrine functions in steroid hormone-regulated tumors. Previous observations had shown that the soluble pattern recognition receptor long pentraxin-3 (PTX3) is a natural selective antagonist for a restricted number of FGF family members, inhibiting FGF2 but not FGF1 and FGF4 activity. Here, we assessed the capacity of PTX3 to antagonize FGF8b and to inhibit the vascularization and growth of steroid hormone-regulated tumors. Surface plasmon resonance analysis shows that PTX3 binds FGF8b with high affinity (K(d) = 30-90 nmol/L). As a consequence, PTX3 prevents the binding of FGF8b to its receptors, inhibits FGF8b-driven ERK1/2 activation, cell proliferation, and chemotaxis in endothelial cells, and suppresses FGF8b-induced neovascularization in vivo. Also, PTX3 inhibits dihydrotestosterone (DHT)- and FGF8b-driven proliferation of androgen-regulated Shionogi 115 (S115) mouse breast tumor cells. Furthermore, DHT-treated, PTX3 overexpressing hPTX3_S115 cell transfectants show a reduced proliferation rate in vitro and a limited angiogenic activity in the chick embryo chorioallantoic membrane and murine s.c. Matrigel plug assays. Accordingly, hPTX3_S115 cells show a dramatic decrease of their tumorigenic activity when grafted in immunodeficient male mice. These results identify PTX3 as a novel FGF8b antagonist endowed with antiangiogenic and antineoplastic activity with possible implications for the therapy of hormonal tumors.
Collapse
Affiliation(s)
- Daria Leali
- Unit of General Pathology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhang K, Brendler E, Geissler A, Fischer S. Synthesis and spectroscopic analysis of cellulose sulfates with regulable total degrees of substitution and sulfation patterns via 13C NMR and FT Raman spectroscopy. POLYMER 2011. [DOI: 10.1016/j.polymer.2010.11.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
37
|
Sangaj N, Kyriakakis P, Yang D, Chang CW, Arya G, Varghese S. Heparin mimicking polymer promotes myogenic differentiation of muscle progenitor cells. Biomacromolecules 2010; 11:3294-300. [PMID: 21058638 PMCID: PMC4136807 DOI: 10.1021/bm101041f] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heparin and heparan sulfate mediated basic fibroblast growth factor (bFGF) signaling plays an important role in skeletal muscle homeostasis by maintaining a balance between proliferation and differentiation of muscle progenitor cells. In this study we investigate the role of a synthetic mimic of heparin, poly(sodium-4-styrenesulfonate) (PSS), on myogenic differentiation of C2C12 cells. Exogenous supplementation of PSS increased the differentiation of C2C12 cells in a dose-dependent manner, while the formation of multinucleated myotubes exhibited a nonmonotonic dependence with the concentration of PSS. Our results further suggest that one possible mechanism by which PSS promotes myogenic differentiation is by downregulating the mitogen activated extracellular regulated signaling kinase (MAPK/ERK) pathway. The binding ability of PSS to bFGF was found to be comparable to heparin through molecular docking calculations and by native PAGE. Such synthetic heparin mimics could offer a cost-effective alternative to heparin and also reduce the risk associated with batch-to-batch variation and contamination of heparin.
Collapse
Affiliation(s)
- Nivedita Sangaj
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Phillip Kyriakakis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Darren Yang
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chien-Wen Chang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gaurav Arya
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shyni Varghese
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
38
|
Antiangiogenic Activity of a Neutralizing Human Single-Chain Antibody Fragment against Fibroblast Growth Factor Receptor 1. Mol Cancer Ther 2010; 9:3244-53. [DOI: 10.1158/1535-7163.mct-10-0417] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Brafman DA, Chang CW, Fernandez A, Willert K, Varghese S, Chien S. Long-term human pluripotent stem cell self-renewal on synthetic polymer surfaces. Biomaterials 2010; 31:9135-44. [PMID: 20817292 DOI: 10.1016/j.biomaterials.2010.08.007] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 08/04/2010] [Indexed: 01/22/2023]
Abstract
Realization of the full potential of human pluripotent stem cells (hPSCs) in regenerative medicine requires the development of well-defined culture conditions for their long-term growth and directed differentiation. Current practices for maintaining hPSCs generally utilize empirically determined combinations of feeder cells and other animal-based products, which are expensive, difficult to isolate, subject to batch-to-batch variations, and unsuitable for cell-based therapies. Using a high-throughput screening approach, we identified several polymers that can support self-renewal of hPSCs. While most of these polymers provide support for only a short period of time, we identified a synthetic polymer poly(methyl vinyl ether-alt-maleic anhydride) (PMVE-alt-MA) that supported the long-term attachment, proliferation and self-renewal of HUES1, HUES9, and iPSCs. The hPSCs cultured on PMVE-alt-MA maintained their characteristic morphology, expressed high levels of markers of pluripotency, and retained a normal karyotype. Such cost-effective, polymer-based matrices that support long-term self-renewal and proliferation of hPSCs will not only help to accelerate the translational perspectives of hPSCs, but also provide a platform to elucidate the underlying molecular mechanisms that regulate stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- David A Brafman
- Department of Bioengineering, University of California-San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0695, United States
| | | | | | | | | | | |
Collapse
|
40
|
Zhang K, Peschel D, Klinger T, Gebauer K, Groth T, Fischer S. Synthesis of carboxyl cellulose sulfate with various contents of regioselectively introduced sulfate and carboxyl groups. Carbohydr Polym 2010. [DOI: 10.1016/j.carbpol.2010.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
41
|
García-Fernández L, Aguilar MR, Fernández MM, Lozano RM, Giménez G, Román JS. Antimitogenic polymer drugs based on AMPS: monomer distribution-bioactivity relationship of water-soluble macromolecules. Biomacromolecules 2010; 11:626-34. [PMID: 20151689 DOI: 10.1021/bm901194e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A number of polysulfonated molecules have demonstrated their interaction with fibroblast growth factor (FGF), hampering their binding to its receptors (low affinity heparan sulfate proteoglycans (HSPG) and high affinity tyrosine kinase FGF receptors) and inhibiting the intracellular signaling and mitogenic response in cultured endothelial cells. The aim of this work was the synthesis and characterization of new copolymers based on 2-acrylamido-2-methylpropane sulfonic acid (AMPS) with antiproliferative activity for antitumoral applications. N-Vinylpyrrolidone (VP) or butyl acrylate (BA) was copolymerized with the sulfonated monomer to obtain macromolecules with different hydrophilic/hydrophobic balance and distribution of the sulfonated groups within the macromolecules. In vitro cell culture proliferative assays showed that monomer distribution affected the inhibition of the proliferative action of FGF. Reactivity ratios of the systems were determined following the free radical copolymerization by in situ (1)H NMR, and the correlation of the monomer sequence distribution with the bioactivity is discussed.
Collapse
Affiliation(s)
- Luis García-Fernández
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Leali D, Bianchi R, Bugatti A, Nicoli S, Mitola S, Ragona L, Tomaselli S, Gallo G, Catello S, Rivieccio V, Zetta L, Presta M. Fibroblast growth factor 2-antagonist activity of a long-pentraxin 3-derived anti-angiogenic pentapeptide. J Cell Mol Med 2010; 14:2109-21. [PMID: 19627396 PMCID: PMC3823002 DOI: 10.1111/j.1582-4934.2009.00855.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fibroblast growth factor-2 (FGF2) plays a major role in angiogenesis. The pattern recognition receptor long-pentraxin 3 (PTX3) inhibits the angiogenic activity of FGF2. To identify novel FGF2-antagonistic peptide(s), four acetylated (Ac) synthetic peptides overlapping the FGF2-binding region PTX3-(97-110) were assessed for their FGF2-binding capacity. Among them, the shortest pentapeptide Ac-ARPCA-NH(2) (PTX3-[100-104]) inhibits the interaction of FGF2 with PTX3 immobilized to a BIAcore sensorchip and suppresses FGF2-dependent proliferation in endothelial cells, without affecting the activity of unrelated mitogens. Also, Ac-ARPCA-NH(2) inhibits angiogenesis triggered by FGF2 or by tumorigenic FGF2-overexpressing murine endothelial cells in chick and zebrafish embryos, respectively. Accordingly, the peptide hampers the binding of FGF2 to Chinese Hamster ovary cells overexpressing the tyrosine-kinase FGF receptor-1 (FGFR1) and to recombinant FGFR1 immobilized to a BIAcore sensorchip without affecting heparin interaction. In all the assays the mutated Ac-ARPSA-NH(2) peptide was ineffective. In keeping with the observation that hydrophobic interactions dominate the interface between FGF2 and the FGF-binding domain of the Ig-like loop D2 of FGFR1, amino acid substitutions in Ac-ARPCA-NH(2) and saturation transfer difference-nuclear magnetic resonance analysis of its mode of interaction with FGF2 implicate the hydrophobic methyl groups of the pentapeptide in FGF2 binding. These results will provide the basis for the design of novel PTX3-derived anti-angiogenic FGF2 antagonists.
Collapse
Affiliation(s)
- Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Peschel D, Zhang K, Aggarwal N, Brendler E, Fischer S, Groth T. Synthesis of novel celluloses derivatives and investigation of their mitogenic activity in the presence and absence of FGF2. Acta Biomater 2010; 6:2116-25. [PMID: 20040386 DOI: 10.1016/j.actbio.2009.12.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 11/24/2009] [Accepted: 12/15/2009] [Indexed: 11/25/2022]
Abstract
Novel cellulose sulfates (CS) with a controlled degree of sulfation (DS(S)) were synthesized through acetosulfation as well as direct sulfation. CS containing carboxyl (CO) or carboxymethyl (CM) groups were prepared by TEMPO oxidation or by carboxymethylation with chloroacetic acid. The derivatization was characterized by nuclear magnetic resonance and Raman spectroscopy. The derivatives were investigated regarding their cytotoxicity and mitogenic activity by modulation of 3T3 fibroblast proliferation with or without exogenous FGF2. All derivatives were non-toxic for 3T3 cells. CS strongly promoted FGF2-induced proliferation, which was positively related to overall DS(S). In the absence of FGF2, minute quantities of CS with intermediate degrees of sulfation exerted stronger mitogenic effects than heparin. No significant promoting effects of CO and CM on cell proliferation were found, though the structure of CO shows similarities to heparin.
Collapse
|
44
|
Rusnati M, Bugatti A, Mitola S, Leali D, Bergese P, Depero LE, Presta M. Exploiting Surface Plasmon Resonance (SPR) Technology for the Identification of Fibroblast Growth Factor-2 (FGF2) Antagonists Endowed with Antiangiogenic Activity. SENSORS (BASEL, SWITZERLAND) 2009; 9:6471-503. [PMID: 22454596 PMCID: PMC3312455 DOI: 10.3390/s90806471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/17/2009] [Accepted: 08/19/2009] [Indexed: 12/31/2022]
Abstract
Angiogenesis, the process of new blood vessel formation, is implicated in various physiological/pathological conditions, including embryonic development, inflammation and tumor growth. Fibroblast growth factor-2 (FGF2) is a heparin-binding angiogenic growth factor involved in various physiopathological processes, including tumor neovascularization. Accordingly, FGF2 is considered a target for antiangiogenic therapies. Thus, numerous natural/synthetic compounds have been tested for their capacity to bind and sequester FGF2 in the extracellular environment preventing its interaction with cellular receptors. We have exploited surface plasmon resonance (SPR) technique in search for antiangiogenic FGF2 binders/antagonists. In this review we will summarize our experience in SPR-based angiogenesis research, with the aim to validate SPR as a first line screening for the identification of antiangiogenic compounds.
Collapse
Affiliation(s)
- Marco Rusnati
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Antonella Bugatti
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Stefania Mitola
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Paolo Bergese
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Laura E. Depero
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| |
Collapse
|
45
|
Christman KL, Vázquez-Dorbatt V, Schopf E, Kolodziej CM, Li RC, Broyer RM, Chen Y, Maynard HD. Nanoscale growth factor patterns by immobilization on a heparin-mimicking polymer. J Am Chem Soc 2008; 130:16585-91. [PMID: 19554729 PMCID: PMC3110987 DOI: 10.1021/ja803676r] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In this study, electrostatic interactions between sulfonate groups of an immobilized polymer and the heparin binding domains of growth factors important in cell signaling were exploited to nanopattern the proteins. Poly(sodium 4-styrenesulfonate-co-poly(ethylene glycol) methacrylate) (pSS-co-pPEGMA) was synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization using ethyl S-thiobenzoyl-2-thiopropionate as a chain transfer agent and 2,2'-azoisobutyronitrile (AIBN) as the initiator. The resulting polymer (1) was characterized by 1H NMR, GPC, FT-IR, and UV-vis and had a number average molecular weight (Mn) of 24,000 and a polydispersity index (PDI) of 1.17. The dithioester end group of 1 was reduced to the thiol, and the polymer was subsequently immobilized on a gold substrate. Binding of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) to the polymer via the heparin binding domains was then confirmed by surface plasmon resonance (SPR). The interactions were stable at physiological salt concentrations. Polymer 1 was cross-linked onto silicon wafers using an electron beam writer forming micro- and nanopatterns. Resolutions of 100 nm and arbitrary nanoscale features such as concentric circles and contiguous squares and triangles were achieved. Fluorescence microscopy confirmed that bFGF and VEGF were subsequently immobilized to the polymer micro- and nanopatterns.
Collapse
Affiliation(s)
- Karen L. Christman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Vimary Vázquez-Dorbatt
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Eric Schopf
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Christopher M. Kolodziej
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Ronald C. Li
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Rebecca M. Broyer
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Yong Chen
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
- California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Dr. East, Los Angeles, CA 90095-1569
| |
Collapse
|
46
|
Polyanionic drugs and viral oncogenesis: a novel approach to control infection, tumor-associated inflammation and angiogenesis. Molecules 2008; 13:2758-85. [PMID: 19002078 PMCID: PMC6245429 DOI: 10.3390/molecules13112758] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 10/13/2008] [Accepted: 10/29/2008] [Indexed: 01/01/2023] Open
Abstract
Polyanionic macromolecules are extremely abundant both in the extracellular environment and inside the cell, where they are readily accessible to many proteins for interactions that play a variety of biological roles. Among polyanions, heparin, heparan sulfate proteoglycans (HSPGs) and glycosphingolipids (GSLs) are widely distributed in biological fluids, at the cell membrane and inside the cell, where they are implicated in several physiological and/or pathological processes such as infectious diseases, angiogenesis and tumor growth. At a molecular level, these processes are mainly mediated by microbial proteins, cytokines and receptors that exert their functions by binding to HSPGs and/or GSLs, suggesting the possibility to use polyanionic antagonists as efficient drugs for the treatment of infectious diseases and cancer. Polysulfated (PS) or polysulfonated (PSN) compounds are a heterogeneous group of natural, semi-synthetic or synthetic molecules whose prototypes are heparin and suramin. Different structural features confer to PS/PSN compounds the capacity to bind and inhibit the biological activities of those same heparin-binding proteins implicated in infectious diseases and cancer. In this review we will discuss the state of the art and the possible future development of polyanionic drugs in the treatment of infectious diseases and cancer.
Collapse
|
47
|
Bronckaers A, Balzarini J, Liekens S. The cytostatic activity of pyrimidine nucleosides is strongly modulated by Mycoplasma hyorhinis infection: Implications for cancer therapy. Biochem Pharmacol 2008; 76:188-97. [PMID: 18555978 DOI: 10.1016/j.bcp.2008.04.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 04/29/2008] [Accepted: 04/30/2008] [Indexed: 02/08/2023]
Abstract
Nucleoside analogues are widely used as chemotherapeutic agents in the treatment of cancer. Several cancers are reported to be associated with mycoplasmas (i.e. Mycoplasma hyorhinis), which contain a number of nucleoside-metabolizing enzymes. Pyrimidine nucleoside analogues, such as 5-fluoro-2'-deoxyuridine (FdUrd), 5-trifluorothymidine (TFT) and 5-halogenated 2'-deoxyuridines can be degraded by thymidine phosphorylase (TP) to their inactive bases. We found in M. hyorhinis-infected MCF-7 breast carcinoma cells (MCF-7/HYOR) a mycoplasma-encoded TP that dramatically (20-150-fold) reduces the cytostatic activity of these compounds. The reduction in cytostatic activity could be fully restored in the presence of TPI (5-chloro-6-[1-(2-iminopyrrolidinyl)methyl]uracil hydrochloride), a known inhibitor of human TP. This observation is in agreement with the markedly decreased formation of active metabolite (i.e. FdUMP for FdUrd) or diminished drug incorporation into nucleic acids (i.e. for TFT and 5-bromo-2'-deoxyuridine) in MCF-7/HYOR cells compared with uninfected MCF-7 cells. Antimetabolite formation is fully restored in the presence of TPI. In contrast, 5-fluoro-5'-deoxyuridine (5'DFUR), an intermediate metabolite of capecitabine, was markedly more cytostatic in MCF-7/HYOR cells than in uninfected cells, due to the activation of this prodrug by the mycoplasma-encoded TP. Thus, our data reveal that M. hyorhinis expresses a TP that activates 5'DFUR but inactivates FdUrd, TFT and 5-halogenated 2'-deoxyuridines, and that is highly sensitive to the inhibitory effect of the TP inhibitor TPI. Given the association of M. hyorhinis with several human cancers, our findings suggest that pyrimidine nucleoside-based but not 5FU-based anti-cancer therapy might be more effective when combined with a mycoplasmal TP inhibitor.
Collapse
|
48
|
Bugatti A, Urbinati C, Ravelli C, De Clercq E, Liekens S, Rusnati M. Heparin-mimicking sulfonic acid polymers as multitarget inhibitors of human immunodeficiency virus type 1 Tat and gp120 proteins. Antimicrob Agents Chemother 2007; 51:2337-45. [PMID: 17452490 PMCID: PMC1913280 DOI: 10.1128/aac.01362-06] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus (HIV) Tat and gp120 intriguingly share the feature of being basic peptides that, once released by HIV(+) cells, bind to polyanionic heparan sulfate proteoglycans (HSPGs) on target uninfected cells, contributing to the onset of AIDS-associated pathologies. To identify multitarget anti-HIV prodrugs, we investigated the gp120 and Tat antagonist potentials of a series of polyanionic synthetic sulfonic acid polymers (SSAPs). Surface plasmon resonance revealed that SSAPs inhibit with a competitive mechanism of action the binding of Tat and gp120 to surface-immobilized heparin, an experimental condition that resembles binding to cellular HSPGs. Accordingly, SSAPs inhibited HSPG-dependent cell internalization and the transactivating activity of Tat. Little is known about the binding of free gp120 to target cells. Here, we identified two classes of gp120 receptors expressed on endothelial cells, one of which was consistent with an HSPG-binding, low-affinity/high-capacity receptor that is inhibited by free heparin. SSAPs inhibited the binding of free gp120 to endothelial cells, as well as its capacity to induce apoptosis in the same cells. In all the assays, poly(4-styrenesulfonic acid) (PSS) proved to be the most potent antagonist of Tat and gp120. Accordingly, PSS bound both proteins with high affinity. In conclusion, SSAPs represent an interesting class of compounds that bind both gp120 and Tat and inhibit their HSPG-dependent cell surface binding and pathological effects. As these activities contribute to both AIDS progression and associated pathologies, SSAPs can be considered prototypic molecules for the development of multitarget drugs for the treatment of HIV infection and AIDS-associated pathologies.
Collapse
Affiliation(s)
- Antonella Bugatti
- General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Liekens S, Gijsbers S, Vanstreels E, Daelemans D, De Clercq E, Hatse S. The nucleotide analog cidofovir suppresses basic fibroblast growth factor (FGF2) expression and signaling and induces apoptosis in FGF2-overexpressing endothelial cells. Mol Pharmacol 2007; 71:695-703. [PMID: 17158200 DOI: 10.1124/mol.106.026559] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cidofovir [(S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine; (S)-HPMPC] is an antiviral drug that has been approved for the treatment of cytomegalovirus retinitis in patients with AIDS. Cidofovir also possesses potent activity against human papillomavirus-induced tumors in animal models and patients. We have recently shown that cidofovir inhibits the development of vascular tumors induced by basic fibroblast growth factor (FGF2)-overexpressing endothelial cells (FGF2-T-MAE) in mice. Here, we demonstrate that the inhibitory activity of cidofovir in FGF2-T-MAE cells may result from the specific induction of apoptosis. Cell cycle analysis revealed that cidofovir induces accumulation of cells in the S phase and, upon prolonged treatment, a significant increase in sub-G1 cells, exhibiting a subdiploid DNA content. Moreover, annexin V binding, an early event in apoptosis induction, was increased in cidofovir-treated FGF2-T-MAE cells. Cidofovir also caused nuclear fragmentation and the activation of caspase-3-like proteases, as evidenced by the cleavage of poly(ADP-ribose)polymerase. In addition, cidofovir treatment of FGF2-T-MAE cells resulted in a pronounced up-regulation of the tumor suppressor protein p53. However, the expression of Bax and Bcl-2 remained unchanged, and cidofovir did not induce the release of cytochrome c from the mitochondria. In addition, cidofovir did not suppress the phosphorylation of protein kinase B/Akt, a transmitter of antiapoptotic survival signals, or its downstream regulator Bad, indicating that the Akt pathway is not affected by cidofovir in FGF2-T-MAE cells. However, the compound inhibited the expression of FGF2 and FGF2 signaling through Erk42/44, as shown by Western blot analysis. Our results indicate that cidofovir inhibits the growth of FGF2-T-MAE cells via inhibition of FGF2 expression and signaling and via the induction of apoptosis. These findings suggest that the clinical use of cidofovir might be expanded to tumors that are not induced by oncogenic viruses.
Collapse
Affiliation(s)
- Sandra Liekens
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
50
|
Hatse S, Balzarini J, Liekens S. Stromal cell-derived factor 1 (CXCL12) binds to endothelial cells and signals through a receptor different from CXCR4. Biochem Biophys Res Commun 2006; 348:192-9. [PMID: 16875673 DOI: 10.1016/j.bbrc.2006.07.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Accepted: 07/09/2006] [Indexed: 11/24/2022]
Abstract
Stromal cell-derived factor 1 (CXCL12) is an angiogenic chemokine that is believed to act solely via its cognate receptor CXCR4. Evidence is now provided for the existence of a different CXCL12 binding and signaling receptor on endothelial cells. Bovine aortic endothelial cells (BAECs) strongly expressed CXCR4 and exhibited high binding capacity for fluorescently labeled CXCL12. However, CXCL12 binding was not correlated with the CXCR4 expression level and was virtually unaffected by the specific CXCR4 antagonists AMD3100 or T22. Similar observations were made in endothelial cells of mouse and human origin. Also, AMD3100 failed to block CXCL12 internalization and CXCL12-induced intracellular signal transduction via extracellular signal-regulated kinases 1/2 in BAECs. In contrast, CXCL12 binding and signaling were almost completely inhibited by the CXCR4 antagonist in T-lymphoid SupT1 cells. Together, our data point to the existence of an additional receptor through which CXCL12 exerts its biological effects in endothelial cells.
Collapse
Affiliation(s)
- Sigrid Hatse
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Minderbroedersstraat 10, B-3000 Leuven, Belgium.
| | | | | |
Collapse
|