Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Polym Chem ; 8(31): 4548-4556, 2017 Aug 20.
Article in English | MEDLINE | ID: mdl-29181096

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.

2.
J Am Chem Soc ; 139(3): 1145-1154, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28079370

ABSTRACT

Many proteins, especially those used as therapeutics, are unstable to storage and shipping temperatures, leading to increased costs in research and industry. Therefore, the design and synthesis of novel stabilizers is an important area of investigation. Herein we report new degradable polymers that stabilize proteins to environmental stressors such as refrigeration and elevated temperature. Specifically, polycaprolactones with different pendant groups were synthesized and surveyed for their ability to stabilize an important therapeutic protein to storage and shipping conditions. Ring-opening polymerization (ROP) of an allyl-substituted caprolactone monomer was carried out using the organocatalyst 1,5,7-triazabicyclo[4.4.0]dec-5-ene (TBD) to yield a well-defined, alkene-substituted degradable polymer, which was used as a common backbone to control for the degree of polymerization. Relevant side chains such as trehalose, lactose, glucose, carboxybetaine, and oligo(ethylene glycol) were installed via postpolymerization thiol-ene reactions. These degradable polymers were then employed as excipients for the stabilization of the therapeutic protein granulocyte colony-stimulating factor (G-CSF) against storage at 4 °C and shipping temperatures of 60 °C. The best stabilization was observed using the trehalose- and zwitterion- substituted polyesters. Both the trehalose- and carboxybetaine-substituted pCL were further investigated with regard to molecular weight dependence, and it was found that the molecular weight was minimally important for stabilization to refrigeration, but critical for G-CSF stabilization at elevated temperatures. Both high performing zwitterionic and trehalose polyesters were also degraded, and the polymers and degradation products were shown to be noncytotoxic. This work provides potential biocompatible polymers for stabilization of the important therapeutic G-CSF, as well as a general platform for the future discovery of new polymeric protein stabilizers.


Subject(s)
Alkenes/chemistry , Granulocyte Colony-Stimulating Factor/chemistry , Polyesters/chemistry , Sulfhydryl Compounds/chemistry , Humans , Models, Molecular , Molecular Conformation , Polyesters/chemical synthesis , Protein Stability
3.
Biomacromolecules ; 17(11): 3417-3440, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27739666

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.


Subject(s)
Anticoagulants/chemistry , Blood Coagulation Disorders/drug therapy , Heparin/chemistry , Polymers/chemistry , Anticoagulants/chemical synthesis , Anticoagulants/therapeutic use , Heparin/chemical synthesis , Heparin/therapeutic use , Humans , Polymers/chemical synthesis , Polymers/therapeutic use
4.
Nanomedicine (Lond) ; 11(20): 2631-2645, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27628768

ABSTRACT

AIM: A promising nanogel vaccine platform was expanded toward antigen conjugation. MATERIALS & METHODS: Block copolymers containing a reactive ester solvophobic block and a PEG-like solvophilic block were synthesized via reversible addition-fragmentation chain-transfer polymerization. Following self-assembly in DMSO, the esters allow for core-crosslinking and hydrophilization by amide bond formation with primary amines. Free thiols were accessed at the polymer chain ends through aminolysis of the reversible addition-fragmentation chain-transfer groups, and into the nanogel core by reactive ester conversion with cysteamine. Subsequently, free thiols were converted into vinyl sulfone moieties. RESULTS: Despite sterical constraints, nanogel-associated vinyl sulfone moieties remained well accessible for cysteins to enforce protein conjugation successfully. CONCLUSION: Our present findings provide a next step toward well-defined vaccine nanoparticles that can co-deliver antigen and a molecular adjuvant.


Subject(s)
Nanocapsules/chemistry , Polymers/chemistry , Sulfones/chemistry , Cross-Linking Reagents/chemistry , Cysteamine/chemistry , Drug Liberation , Esters/chemical synthesis , Esters/chemistry , Gels , Humans , Polyethylene Glycols/chemistry , Polymers/chemical synthesis , Serum Albumin, Bovine/chemistry , Sulfhydryl Compounds/chemistry , Surface Properties
5.
Biomacromolecules ; 17(10): 3386-3395, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27580376

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.


Subject(s)
Fibroblast Growth Factor 2/chemistry , Heparitin Sulfate/chemistry , Methacrylates/chemistry , Polyethylene Glycols/chemistry , Cell Line , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/pharmacology , Heparitin Sulfate/pharmacology , Humans , Methacrylates/pharmacology , Polyethylene Glycols/pharmacology , Polymers/chemistry , Polymers/pharmacology , Regeneration/drug effects , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology , Wound Healing/drug effects
6.
Langmuir ; 32(16): 4043-51, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27078573

ABSTRACT

Electron beam (e-beam) lithography was employed to prepare one protein immobilized hydrogel encapsulated inside another by first fabricating protein-reactive hydrogels of orthogonal reactivity and subsequently conjugating the biomolecules. Exposure of thin films of eight arm star poly(ethylene glycol) (PEG) functionalized with biotin (Biotin-PEG), alkyne (Alkyne-PEG) or aminooxy (AO-PEG) end-groups to e-beam radiation resulted in cross-linked hydrogels with the respective functionality. It was determined via confocal microscopy that a nominal size exclusion effect exists for streptavidin immobilized on Biotin-PEG hydrogels of feature sizes ranging from 5 to 40 µm. AO-PEG was subsequently patterned as an encapsulated core inside a contiguous outer shell of Biotin-PEG. Similarly, Alkyne-PEG was patterned as a core inside an AO-PEG shell. The hydrogel reactive end-groups were conjugated to dyes or proteins of complementary reactivity, and the three-dimensional (3-D) spatial orientation was determined for both configurations using confocal microscopy. The enzyme glucose oxidase (GOX) was immobilized in the core of the encapsulated Alkyne-PEG core/ AO-PEG shell architecture, and horseradish peroxidase (HRP) was conjugated to the shell periphery. Bioactivity for the HRP-GOX enzyme pair was observed in this encapsulated configuration by demonstrating that the enzyme pair was capable of enzyme cascade reactions.


Subject(s)
Electrons , Horseradish Peroxidase/metabolism , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Printing , Alkynes/chemistry , Biotin/chemistry , Capsules , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism
7.
Macromolecules ; 49(1): 30-37, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26949271

ABSTRACT

Tetrazine end-functionalized telechelic polymers were synthesized by controlled radical polymerization (CRP) and employed to generate T4 Lysozyme homodimers. Mutant T4 Lysozyme (V131C), containing a single surface-exposed cysteine, was modified with a protein-reactive trans-cyclooctene (T4L-TCO). Reversible addition-fragmentation chain transfer (RAFT) polymerization yielded poly(N-isopropylacrylamide) (pNIPAAm) with a number average molecular weight (Mn by 1H-NMR) of 2.0 kDa and a dispersity (D by GPC) of 1.05. pNIPAAm was then modified at both ends by post-polymerization with 6-methyl tetrazine. For comparison, 2.0 kDa bis-tetrazine poly(ethylene glycol) (PEG) and 2.0 kDa bis-maleimide pNIPAAm were synthesized. Ligation of T4L-TCO to bis-tetrazine pNIPAAm or bis-tetrazine PEG resulted in protein homodimer in 38% yield and 37% yield, respectively, after only 1 hour, whereas bis-maleimide pNIPAAm resulted in only 5% yield of dimer after 24 h. This work illustrates the advantage of employing tetrazine ligation over maleimide thiol-ene chemistry for the synthesis of protein homodimer conjugates.

8.
Biomaterials ; 81: 157-168, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26731578

ABSTRACT

Site-specific chemical dimerization of fibroblast growth factor 2 (FGF2) with the optimal linker length resulted in a FGF2 homodimer with improved granulation tissue formation and blood vessel formation at exceptionally low concentrations. Homodimers of FGF2 were synthesized through site-specific linkages to both ends of different molecular weight poly(ethylene glycols) (PEGs). The optimal linker length was determined by screening dimer-induced metabolic activity of human dermal fibroblasts and found to be that closest to the inter-cysteine distance, 70 Å, corresponding to 2 kDa PEG. A straightforward analysis of the kinetics of second ligand binding as a function of tether length showed that, as the polymerization index (the number of monomer repeat units in the polymer, N) of the tether decreases, the mean time for second ligand capture decreases as ∼N(3/2), leading to an enhancement of the number of doubly bound ligands in steady-state for a given (tethered) ligand concentration. FGF2-PEG2k-FGF2 induced greater fibroblast metabolic activity than FGF2 alone, all other dimers, and all monoconjugates, at each concentration tested, with the greatest difference observed at low (0.1 ng/mL) concentration. FGF2-PEG2k-FGF2 further exhibited superior activity compared to FGF2 for both metabolic activity and migration in human umbilical vein endothelial cells, as well as improved angiogenesis in a coculture model in vitro. Efficacy in an in vivo wound healing model was assessed in diabetic mice. FGF2-PEG2k-FGF2 increased granulation tissue and blood vessel density in the wound bed compared to FGF2. The results suggest that this rationally designed construct may be useful for improving the fibroblast matrix formation and angiogenesis in chronic wound healing.


Subject(s)
Fibroblast Growth Factor 2/agonists , Fibroblast Growth Factor 2/pharmacology , Granulation Tissue/pathology , Protein Multimerization , Wound Healing/drug effects , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Blotting, Western , Cell Movement/drug effects , Cross-Linking Reagents/chemistry , Dermis/cytology , Diabetes Mellitus, Experimental/pathology , Fibroblast Growth Factor 2/chemistry , Fibroblasts/cytology , Fibroblasts/drug effects , Granulation Tissue/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Male , Mice , Neovascularization, Physiologic/drug effects , Polyethylene Glycols/chemistry
9.
Biomacromolecules ; 16(9): 2684-92, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26212474

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.


Subject(s)
Biomimetic Materials , Fibroblast Growth Factor 2 , Polyvinyls , Sulfonic Acids , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Heparin/chemistry , Humans , Polyvinyls/chemistry , Polyvinyls/pharmacology , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...