Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Article En | MEDLINE | ID: mdl-38613808

Glycosaminoglycans (GAGs) are ubiquitous components in the cartilage extracellular matrix (ECM). Ultrastructural arrangement of ECM and GAG-mediated interactions with collagen are known to govern the mechanics in articular cartilage, but these interactions are less clear in other cartilage types. Therefore, this article reviews the current literature on ultrastructure of articular, auricular, meniscal, and nasal septal cartilage, seeking insight into GAG-mediated interactions influencing mechanics. Ultrastructural features of these cartilages are discussed to highlight differences between them. GAG-mediated interactions are reviewed under two categories: interactions with chondrocytes and interactions with other fibrillar macromolecules of the ECM. Moreover, efforts to replicate GAG-mediated interactions to improve mechanical integrity of tissue-engineered cartilage constructs are discussed. In conclusion, studies exploring cartilage specific GAGs are poorly represented in the literature, and the ultrastructure of nasal septal and auricular cartilage is less studied compared with articular and meniscal cartilages. Understanding the contribution of GAGs in cartilage mechanics at the ultrastructural level and translating that knowledge to engineered cartilage will facilitate improvement of cartilage tissue engineering approaches.

2.
Biomater Sci ; 12(1): 134-150, 2023 Dec 19.
Article En | MEDLINE | ID: mdl-37933486

Synthetic polymers, such as poly(vinyl alcohol) (PVA), are popular biomaterials for the fabrication of hydrogels for tissue engineering and regenerative medicine (TERM) applications, as they provide excellent control over the physico-chemical properties of the hydrogel. However, their bioinert nature is known to limit cell-biomaterial interactions by hindering cell infiltration, blood vessel recruitment and potentially limiting their integration with the host tissue. Efforts in the field have therefore focused on increasing the biofunctionality of synthetic hydrogels, without limiting the advantages associated with their tailorability and controlled release capacity. The aim of this study was to investigate the suitability of pristine gelatin to enhance the biofunctionality of tyraminated PVA (PVA-Tyr) hydrogels, by promoting cell infiltration and host blood vessel recruitment for TERM applications. Pure PVA-Tyr hydrogels and PVA-Tyr hydrogels incorporated with vascular endothelial growth factor (VEGF), a well-known pro-angiogenic stimulus, were used for comparison. Incorporating increasing concentrations of VEGF (0.01-10 µg mL-1) or gelatin (0.01-5 wt%) did not influence the physical properties of PVA-Tyr hydrogels. However, their presence within the polymer network (>0.1 µg mL-1 VEGF and >0.1 wt% gelatin) promoted endothelial cell interactions with the hydrogels. The covalent binding of unmodified gelatin or VEGF to the PVA-Tyr network did not hamper their inherent bioactivity, as they both promoted angiogenesis in a chick chorioallantoic membrane (CAM) assay, performing comparably with the unbound VEGF control. When the PVA-Tyr hydrogels were implanted subcutaneously in mice, it was observed that cell infiltration into the hydrogels was possible in the absence of gelatin or VEGF at 1- or 3-weeks post-implantation, highlighting a clear difference between in vitro an in vivo cell-biomaterial interaction. Nevertheless, the presence of gelatin or VEGF was necessary to enhance blood vessel recruitment and infiltration, although no significant difference was observed between these two biological molecules. Overall, this study highlights the potential of gelatin as a standalone pro-angiogenic cue to enhance biofunctionality of synthetic hydrogels and provides promise for their use in a variety of TERM applications.


Polyvinyl Alcohol , Vascular Endothelial Growth Factor A , Mice , Animals , Polyvinyl Alcohol/chemistry , Gelatin/chemistry , Tissue Engineering , Hydrogels/chemistry , Polymers/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Ethanol
3.
Int J Mol Sci ; 24(18)2023 Sep 14.
Article En | MEDLINE | ID: mdl-37762403

This review examines the roles of HS-proteoglycans (HS-PGs) in general, and, in particular, perlecan and syndecan as representative examples and their interactive ligands, which regulate physiological processes and cellular behavior in health and disease. HS-PGs are essential for the functional properties of tissues both in development and in the extracellular matrix (ECM) remodeling that occurs in response to trauma or disease. HS-PGs interact with a biodiverse range of chemokines, chemokine receptors, protease inhibitors, and growth factors in immune regulation, inflammation, ECM stabilization, and tissue protection. Some cell regulatory proteoglycan receptors are dually modified hybrid HS/CS proteoglycans (betaglycan, CD47). Neurexins provide synaptic stabilization, plasticity, and specificity of interaction, promoting neurotransduction, neurogenesis, and differentiation. Ternary complexes of glypican-1 and Robbo-Slit neuroregulatory proteins direct axonogenesis and neural network formation. Specific neurexin-neuroligin complexes stabilize synaptic interactions and neural activity. Disruption in these interactions leads to neurological deficits in disorders of functional cognitive decline. Interactions with HS-PGs also promote or inhibit tumor development. Thus, HS-PGs have complex and diverse regulatory roles in the physiological processes that regulate cellular behavior and the functional properties of normal and pathological tissues. Specialized HS-PGs, such as the neurexins, pikachurin, and Eyes-shut, provide synaptic stabilization and specificity of neural transduction and also stabilize the axenome primary cilium of phototoreceptors and ribbon synapse interactions with bipolar neurons of retinal neural networks, which are essential in ocular vision. Pikachurin and Eyes-Shut interactions with an α-dystroglycan stabilize the photoreceptor synapse. Novel regulatory roles for HS-PGs controlling cell behavior and tissue function are expected to continue to be uncovered in this fascinating class of proteoglycan.


Heparan Sulfate Proteoglycans , Physiological Phenomena , Glycosaminoglycans , Glypicans , Syndecans
4.
Am J Physiol Cell Physiol ; 324(1): C142-C152, 2023 01 01.
Article En | MEDLINE | ID: mdl-36409173

Chondroitin sulfate (CS) is a ubiquitous glycosaminoglycan covalently attached to the core proteins of cell surface, extracellular, and intracellular proteoglycans. The multistep and highly regulated biosynthesis of chondroitin sulfate and its degradation products give rise to a diverse species of molecules with functional regulatory properties in biological systems. This review will elucidate and expand on the most recent advances in understanding the role of chondroitin sulfate and its associate proteoglycans, in arthritis and Duchenne muscular dystrophy (DMD), two different and discrete pathologies. Highlighting not only the biodiverse nature of this family of molecules but also the utilization of CS proteoglycans, CS, and its catabolic fragments as biomarkers and potential therapeutic targets for disease pathologies.


Arthritis , Muscular Dystrophy, Duchenne , Humans , Proteoglycans/metabolism , Chondroitin Sulfates/metabolism , Muscular Dystrophy, Duchenne/metabolism , Chondroitin Sulfate Proteoglycans
5.
Front Cell Dev Biol ; 10: 856261, 2022.
Article En | MEDLINE | ID: mdl-35433700

This review highlights the multifunctional properties of perlecan (HSPG2) and its potential roles in repair biology. Perlecan is ubiquitous, occurring in vascular, cartilaginous, adipose, lymphoreticular, bone and bone marrow stroma and in neural tissues. Perlecan has roles in angiogenesis, tissue development and extracellular matrix stabilization in mature weight bearing and tensional tissues. Perlecan contributes to mechanosensory properties in cartilage through pericellular interactions with fibrillin-1, type IV, V, VI and XI collagen and elastin. Perlecan domain I - FGF, PDGF, VEGF and BMP interactions promote embryonic cellular proliferation, differentiation, and tissue development. Perlecan domain II, an LDLR-like domain interacts with lipids, Wnt and Hedgehog morphogens. Perlecan domain III binds FGF-7 and 18 and has roles in the secretion of perlecan. Perlecan domain IV, an immunoglobulin repeat domain, has cell attachment and matrix stabilizing properties. Perlecan domain V promotes tissue repair through interactions with VEGF, VEGF-R2 and α2ß1 integrin. Perlecan domain-V LG1-LG2 and LG3 fragments antagonize these interactions. Perlecan domain V promotes reconstitution of the blood brain barrier damaged by ischemic stroke and is neurogenic and neuroprotective. Perlecan-VEGF-VEGFR2, perlecan-FGF-2 and perlecan-PDGF interactions promote angiogenesis and wound healing. Perlecan domain I, III and V interactions with platelet factor-4 and megakaryocyte and platelet inhibitory receptor promote adhesion of cells to implants and scaffolds in vascular repair. Perlecan localizes acetylcholinesterase in the neuromuscular junction and is of functional significance in neuromuscular control. Perlecan mutation leads to Schwartz-Jampel Syndrome, functional impairment of the biomechanical properties of the intervertebral disc, variable levels of chondroplasia and myotonia. A greater understanding of the functional working of the neuromuscular junction may be insightful in therapeutic approaches in the treatment of neuromuscular disorders. Tissue engineering of salivary glands has been undertaken using bioactive peptides (TWSKV) derived from perlecan domain IV. Perlecan TWSKV peptide induces differentiation of salivary gland cells into self-assembling acini-like structures that express salivary gland biomarkers and secrete α-amylase. Perlecan also promotes chondroprogenitor stem cell maturation and development of pluripotent migratory stem cell lineages, which participate in diarthrodial joint formation, and early cartilage development. Recent studies have also shown that perlecan is prominently expressed during repair of adult human articular cartilage. Perlecan also has roles in endochondral ossification and bone development. Perlecan domain I hydrogels been used in tissue engineering to establish heparin binding growth factor gradients that promote cell migration and cartilage repair. Perlecan domain I collagen I fibril scaffolds have also been used as an FGF-2 delivery system for tissue repair. With the availability of recombinant perlecan domains, the development of other tissue repair strategies should emerge in the near future. Perlecan co-localization with vascular elastin in the intima, acts as a blood shear-flow endothelial sensor that regulates blood volume and pressure and has a similar role to perlecan in canalicular fluid, regulating bone development and remodeling. This complements perlecan's roles in growth plate cartilage and in endochondral ossification to form the appendicular and axial skeleton. Perlecan is thus a ubiquitous, multifunctional, and pleomorphic molecule of considerable biological importance. A greater understanding of its diverse biological roles and functional repertoires during tissue development, growth and disease will yield valuable insights into how this impressive proteoglycan could be utilized successfully in repair biology.

6.
Adv Healthc Mater ; 10(14): e2100388, 2021 07.
Article En | MEDLINE | ID: mdl-33890424

Surface modification of biomaterials is a promising approach to control biofunctionality while retaining the bulk biomaterial properties. Perlecan is the major proteoglycan in the vascular basement membrane that supports low levels of platelet adhesion but not activation. Thus, perlecan is a promising bioactive for blood-contacting applications. This study furthers the mechanistic understanding of platelet interactions with perlecan by establishing that platelets utilize domains III and V of the core protein for adhesion. Polyvinyl chloride (PVC) is functionalized with recombinant human perlecan domain V (rDV) to explore the effect of the tethering method on proteoglycan orientation and bioactivity. Tethering of rDV to PVC is achieved via either physisorption or covalent attachment via plasma immersion ion implantation (PIII) treatment. Both methods of rDV tethering reduce platelet adhesion and activation compared to the pristine PVC, however, the mechanisms are unique for each tethering method. Physisorption of rDV on PVC orientates the molecule to hinder access to the integrin-binding region, which inhibits platelet adhesion. In contrast, PIII treatment orientates rDV to allow access to the integrin-binding region, which is rendered antiadhesive to platelets via the glycosaminoglycan (GAG) chain. These effects demonstrate the potential of rDV biofunctionalization to modulate platelet interactions for blood contacting applications.


Heparan Sulfate Proteoglycans , Polyvinyl Chloride , Extracellular Matrix Proteins , Glycosaminoglycans , Humans
7.
Cartilage ; 13(2_suppl): 476S-485S, 2021 12.
Article En | MEDLINE | ID: mdl-33749320

OBJECTIVE: To investigate GAG-ECM (glycosaminoglycan-extracellular matrix) interactions in different cartilage types. To achieve this, we first aimed to determine protocols for consistent calculation of GAG content between cartilage types. DESIGN: Auricular cartilage containing both collagen and elastin was used to determine the effect of lyophilization on GAG depletion activity. Bovine articular, auricular, meniscal, and nasal cartilage plugs were treated using different reagents to selectively remove GAGs. Sulfated glycosaminoglycan (sGAG) remaining in the sample after treatment were measured, and sGAG loss was compared between cartilage types. RESULTS: The results indicate that dry weight of cartilage should be measured prior to cartilage treatment in order to provide a more accurate reference for normalization. Articular, meniscal, and nasal cartilage lost significant amounts of sGAG for all reagents used. However, only hyaluronidase was able to remove significant amount of sGAG from auricular cartilage. Furthermore, hyaluronidase was able to remove over 99% of sGAG from all cartilage types except auricular cartilage where it only removed around 76% of sGAG. The results indicate GAG-specific ECM binding for different cartilage types and locations. CONCLUSIONS: In conclusion, lyophilization can be performed to determine native dry weight for normalization without affecting the degree of GAG treatment. To our knowledge, this is the first study to compare GAG-ECM interactions of different cartilage types using different GAG extraction methods. Degree of GAG depletion not only varied with cartilage type but also the same type from different anatomic locations. This suggests specific structure-function roles for GAG populations found in the tissues.


Cartilage , Meniscus , Animals , Cartilage/metabolism , Cattle , Collagen/metabolism , Extracellular Matrix/metabolism , Meniscus/metabolism
8.
Biomacromolecules ; 22(4): 1590-1599, 2021 04 12.
Article En | MEDLINE | ID: mdl-33764748

A rapid photo-curing system based on poly(2-ethyl-2-oxazoline-co-2-allylamidopropyl-2-oxazoline) and its in vivo compatibility are presented. The base polymer was synthesized from the copolymerization of 2-ethyl-2-oxazoline (EtOx) and the methyl ester containing 2-methoxycarboxypropyl-2-oxazoline (C3MestOx) followed by amidation with allylamine to yield a highly water-soluble macromer. We showed that spherical hydrogels can be obtained by a simple water-in-oil gelation method using thiol-ene coupling and investigated the in vivo biocompatibility of these hydrogel spheres in a 28-day murine subdermal model. For comparison, hydrogel spheres prepared from poly(ethylene glycol) were also implanted. Both materials displayed mild, yet typical foreign body responses with little signs of fibrosis. This is the first report on the foreign body response of a poly(2-oxazoline) hydrogel, which paves the way for future investigations into how this highly tailorable class of materials can be used for implantable hydrogel devices.


Hydrogels , Polyethylene Glycols , Animals , Kinetics , Mice , Polymerization , Polymers
9.
J Funct Biomater ; 10(3)2019 Jul 30.
Article En | MEDLINE | ID: mdl-31366056

Poly(d,l-lactide-co-glycolide) (PLGA) has been extensively explored for bone regeneration applications; however, its clinical use is limited by low osteointegration. Therefore, approaches that incorporate osteoconductive molecules are of great interest. Graphene oxide (GO) is gaining popularity for biomedical applications due to its ability to bind biological molecules and present them for enhanced bioactivity. This study reports the preparation of PLGA microparticles via Pickering emulsification using GO as the sole surfactant, which resulted in hybrid microparticles in the size range of 1.1 to 2.4 µm based on the ratio of GO to PLGA in the reaction. Furthermore, this study demonstrated that the hybrid GO-PLGA microparticles were not cytotoxic to either primary human fetal cartilage rudiment cells or the human osteoblast-like cell line, Saos-2. Additionally, the GO-PLGA microparticles promoted the osteogenic differentiation of the human fetal cartilage rudiment cells in the absence of exogenous growth factors to a greater extent than PLGA alone. These findings demonstrate that GO-PLGA microparticles are cytocompatible, osteoinductive and have potential as substrates for bone tissue engineering.

10.
J Biol Chem ; 294(30): 11458-11472, 2019 07 26.
Article En | MEDLINE | ID: mdl-31175155

Mast cells represent a heterogeneous cell population that is well-known for the production of heparin and the release of histamine upon activation. Serglycin is a proteoglycan that within mast cell α-granules is predominantly decorated with the glycosaminoglycans heparin or chondroitin sulfate (CS) and has a known role in granule homeostasis. Heparanase is a heparin-degrading enzyme, is present within the α-granules, and contributes to granule homeostasis, but an equivalent CS-degrading enzyme has not been reported previously. In this study, using several approaches, including epitope-specific antibodies, immunohistochemistry, and EM analyses, we demonstrate that human HMC-1 mast cells produce the CS-degrading enzymes hyaluronidase-1 (HYAL1) and HYAL4. We observed that treating the two model CS proteoglycans aggrecan and serglycin with HYAL1 and HYAL4 in vitro cleaves the CS chains into lower molecular weight forms with nonreducing end oligosaccharide structures similar to CS stub neoepitopes generated after digestion with the bacterial lyase chondroitinase ABC. We found that these structures are associated with both the CS linkage region and with structures more distal toward the nonreducing end of the CS chain. Furthermore, we noted that HYAL4 cleaves CS chains into lower molecular weight forms that range in length from tetra- to dodecasaccharides. These results provide first evidence that mast cells produce HYAL4 and that this enzyme may play a specific role in maintaining α-granule homeostasis in these cells by cleaving CS glycosaminoglycan chains attached to serglycin.


Chondroitin Sulfates/metabolism , Hyaluronoglucosaminidase/biosynthesis , Mast Cells/enzymology , Proteoglycans/metabolism , Vesicular Transport Proteins/metabolism , Aggrecans/chemistry , Aggrecans/metabolism , Animals , Chondroitin Sulfates/chemistry , Humans , Molecular Weight , Proteoglycans/chemistry , Vesicular Transport Proteins/chemistry
11.
Front Oncol ; 9: 1482, 2019.
Article En | MEDLINE | ID: mdl-32010611

Cancer metastasis is the dissemination of tumor cells to new sites, resulting in the formation of secondary tumors. This process is complex and is spatially and temporally regulated by intrinsic and extrinsic factors. One important extrinsic factor is the extracellular matrix, the non-cellular component of tissues. Heparan sulfate proteoglycans (HSPGs) are constituents of the extracellular matrix, and through their heparan sulfate chains and protein core, modulate multiple events that occur during the metastatic cascade. This review will provide an overview of the role of the extracellular matrix in the events that occur during cancer metastasis, primarily focusing on perlecan. Perlecan, a basement membrane HSPG is a key component of the vascular extracellular matrix and is commonly associated with events that occur during the metastatic cascade. Its contradictory role in these events will be discussed and we will highlight the recent advances in cancer therapies that target HSPGs and their modifying enzymes.

12.
J Histochem Cytochem ; 66(4): 321-336, 2018 04.
Article En | MEDLINE | ID: mdl-29290153

Key events that occur during inflammation include the recruitment, adhesion, and transmigration of leukocytes from the circulation to the site of inflammation. These events are modulated by chemokines, integrins, and selectins and the interaction of these molecules with glycosaminoglycans, predominantly heparan sulfate (HS). The development of HS/heparin mimetics that interfere or inhibit the interactions that occur between glycosaminoglycans and modulators of inflammation holds great potential for use as anti-inflammatory therapeutics. This review will detail the role of HS in the events that occur during inflammation, their interaction and modulation of inflammatory mediators, and the current advances in the development of HS/heparin mimetics as anti-inflammatory biotherapeutics.


Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/pharmacology , Drug Discovery , Heparitin Sulfate/immunology , Heparitin Sulfate/pharmacology , Immunity, Innate , Inflammation/immunology , Animals , Anti-Inflammatory Agents/chemistry , Biomimetics/methods , Chemokines/immunology , Drug Discovery/methods , Glucuronidase/antagonists & inhibitors , Glucuronidase/immunology , Heparitin Sulfate/chemistry , Humans , Immunity, Innate/drug effects , Inflammation/drug therapy
13.
Macromol Biosci ; 17(12)2017 12.
Article En | MEDLINE | ID: mdl-29068543

Multicomponent gelatin-methacryloyl (GelMA) hydrogels are regularly adopted for cartilage tissue engineering (TE) applications, where optimizing chemical modifications for preserving biofunctionality is often overlooked. This study investigates the biological effect of two different modification methods, methacrylation and thiolation, to copolymerize GelMA and heparin. The native bioactivity of methacrylated heparin (HepMA) and thiolated heparin (HepSH) is evaluated via thromboplastin time and heparan sulfate-deficient myeloid cell-line proliferation assay, demonstrating that thiolation is superior for preserving anticoagulation and growth factor signaling capacity. Furthermore, incorporating either HepMA or HepSH in chondrocyte-laden GelMA hydrogels, cultured for 5 weeks under chondrogenic conditions, promotes cell viability and chondrocyte phenotype. However, only GelMA-HepSH hydrogels yield significantly greater differentiation and matrix deposition in vitro compared to GelMA. This study demonstrates that thiol-ene chemistry offers a favorable strategy for incorporating bioactives into gelatin hydrogels as compared to methacrylation while furthermore highlighting GelMA-HepSH hydrogels as candidates for cartilage TE applications.


Chondrocytes/cytology , Chondrogenesis/drug effects , Heparin/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Cartilage, Articular/cytology , Cell Differentiation/drug effects , Cell Survival/drug effects , Chondrocytes/drug effects , Extracellular Matrix , Fibroblast Growth Factor 2/metabolism , Gelatin/chemistry , Gelatin/pharmacology , Heparin/chemistry , Heparin/metabolism , Humans , Methacrylates/chemistry , Partial Thromboplastin Time , Photochemistry/methods , Sulfhydryl Compounds/chemistry , Tissue Engineering/methods , Tissue Scaffolds
14.
J Control Release ; 250: 48-61, 2017 03 28.
Article En | MEDLINE | ID: mdl-28189628

The repair of dermal wounds, particularly in the diabetic population, poses a significant healthcare burden. The impaired wound healing of diabetic wounds is attributed to low levels of endogenous growth factors, including vascular endothelial growth factor (VEGF), that normally stimulate multiple phases of wound healing. In this study, chitosan scaffolds were prepared via freeze drying and loaded with plasmid DNA encoding perlecan domain I and VEGF189 and analyzed in vivo for their ability to promote dermal wound healing. The plasmid DNA encoding perlecan domain I and VEGF189 loaded scaffolds promoted dermal wound healing in normal and diabetic rats. This treatment resulted in an increase in the number of blood vessels and sub-epithelial connective tissue matrix components within the wound beds compared to wounds treated with chitosan scaffolds containing control DNA or wounded controls. These results suggest that chitosan scaffolds containing plasmid DNA encoding VEGF189 and perlecan domain I have the potential to induce angiogenesis and wound healing.


Chitosan/chemistry , DNA/administration & dosage , Heparan Sulfate Proteoglycans/genetics , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/genetics , Wound Healing , Animals , DNA/chemistry , Diabetes Complications/physiopathology , Drug Delivery Systems , Drug Liberation , Heparan Sulfate Proteoglycans/metabolism , Humans , Male , Mechanical Phenomena , Plasmids , Rats, Inbred Lew , Skin/blood supply , Skin/injuries , Tissue Scaffolds , Transgenes , Vascular Endothelial Growth Factor A/metabolism
15.
J Biol Chem ; 292(10): 4054-4063, 2017 03 10.
Article En | MEDLINE | ID: mdl-28115521

Platelet factor 4 (PF4) is produced by platelets with roles in both inflammation and wound healing. PF4 is stored in platelet α-granules bound to the glycosaminoglycan (GAG) chains of serglycin. This study revealed that platelet serglycin is decorated with chondroitin/dermatan sulfate and that PF4 binds to these GAG chains. Additionally, PF4 had a higher affinity for endothelial-derived perlecan heparan sulfate chains than serglycin GAG chains. The binding of PF4 to perlecan was found to inhibit both FGF2 signaling and platelet activation. This study revealed additional insight into the ways in which PF4 interacts with components of the vasculature to modulate cellular events.


Blood Platelets/metabolism , Chondroitin Sulfates/metabolism , Dermatan Sulfate/metabolism , Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Platelet Factor 4/metabolism , Proteoglycans/metabolism , Vesicular Transport Proteins/metabolism , Blotting, Western , Humans , Platelet Activation , Protein Binding
16.
J Tissue Eng ; 7: 2041731416677132, 2016.
Article En | MEDLINE | ID: mdl-27895888

Heparin-based hydrogels are attractive for controlled growth factor delivery, due to the native ability of heparin to bind and stabilize growth factors. Basic fibroblast growth factor and vascular endothelial growth factor are heparin-binding growth factors that synergistically enhance angiogenesis. Mild, in situ encapsulation of both basic fibroblast growth factor and vascular endothelial growth factor and subsequent bioactive dual release has not been demonstrated from heparin-crosslinked hydrogels, and the combined long-term delivery of both growth factors from biomaterials is still a major challenge. Both basic fibroblast growth factor and vascular endothelial growth factor were encapsulated in poly(vinyl alcohol)-heparin hydrogels and demonstrated controlled release. A model cell line, BaF32, was used to show bioactivity of heparin and basic fibroblast growth factor released from the gels over multiple days. Released basic fibroblast growth factor promoted higher human umbilical vein endothelial cell outgrowth over 24 h and proliferation for 3 days than the poly(vinyl alcohol)-heparin hydrogels alone. The release of vascular endothelial growth factor from poly(vinyl alcohol)-heparin hydrogels promoted human umbilical vein endothelial cell outgrowth but not significant proliferation. Dual-growth factor release of basic fibroblast growth factor and vascular endothelial growth factor from poly(vinyl alcohol)-heparin hydrogels resulted in a synergistic effect with significantly higher human umbilical vein endothelial cell outgrowth compared to basic fibroblast growth factor or vascular endothelial growth factor alone. Poly(vinyl alcohol)-heparin hydrogels allowed bioactive growth factor encapsulation and provided controlled release of multiple growth factors which is beneficial toward tissue regeneration applications.

17.
Biointerphases ; 11(2): 029701, 2016 06 27.
Article En | MEDLINE | ID: mdl-27233532

Platelets are routinely stored enabling transfusions for a range of conditions. While the current platelet storage bags, composed of either polyvinylchloride or polyolefin, are well-established, the storage of platelets in these bags beyond 7 days reduces platelet viability below clinically usable levels. New materials and coatings that promote platelet respiration while not supporting platelet adhesion or activation have started to emerge, with the potential to enable platelet storage beyond 7 days. This review focuses on the literature describing currently used biomaterials for platelet storage and emerging materials that are showing promise for improving platelet storage.


Blood Platelets/physiology , Coated Materials, Biocompatible , Preservation, Biological/methods , Cell Survival , Humans , Platelet Transfusion
18.
J Biomed Mater Res A ; 104(7): 1736-46, 2016 07.
Article En | MEDLINE | ID: mdl-26946213

Cerium oxide nanoparticles are being widely explored for cell therapies. In this study, nanoceria was functionalized with hyaluronan (HA) using the organosilane linker, 3-aminopropyltriethoxysilane. HA-nanoceria was found to be cytocompatible and to reduce intracellular reactive oxygen species in human fibroblasts. The HA-nanoceria was found to colocalize with CD44 on the surface of the cells and once internalized traffic to the lysosomes, be degraded and induce markers of autophagy. These particles were also effective in reducing the cell surface expression of CD44. Together these data suggest that HA-nanoceria is a promising drug delivery material to target CD44-expressing cells through a variety of mechanisms. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1736-1746, 2016.


Cerium/chemistry , Coated Materials, Biocompatible/pharmacology , Fibroblasts/metabolism , Hyaluronan Receptors/metabolism , Hyaluronic Acid/pharmacology , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , Cell Communication/drug effects , Cell Line , Cell Proliferation/drug effects , Fetus/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Flow Cytometry , Humans , Intracellular Space/metabolism , Lung/cytology , Lysosomes/drug effects , Lysosomes/metabolism , Nanoparticles/ultrastructure , Spectroscopy, Fourier Transform Infrared , Thermogravimetry
19.
J Histochem Cytochem ; 64(2): 85-98, 2016 02.
Article En | MEDLINE | ID: mdl-26586669

The granules of mast cells contain a myriad of mediators that are stored and protected by the sulfated glycosaminoglycan (GAG) chains that decorate proteoglycans. Whereas heparin is the GAG predominantly associated with mast cells, mast cell proteoglycans are also decorated with heparan sulfate and chondroitin sulfate (CS). This study investigated a unique CS structure produced by mast cells that was detected with the antibody clone 2B6 in the absence of chondroitinase ABC digestion. Mast cells in rodent tissue sections were characterized using toluidine blue, Leder stain and the presence of mast cell tryptase. The novel CS epitope was identified in rodent tissue sections and localized to cells that were morphologically similar to cells chemically identified as mast cells. The rodent mast cell-like line RBL-2H3 was also shown to express the novel CS epitope. This epitope co-localized with multiple CS proteoglycans in both rodent tissue and RBL-2H3 cultured cells. These findings suggest that the novel CS epitope that decorates mast cell proteoglycans may play a role in the way these chains are structured in mast cells.


Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/immunology , Mast Cells/cytology , Mast Cells/immunology , Animals , Chondroitin Sulfate Proteoglycans/analysis , Chondroitin Sulfates/analysis , Epitope Mapping , Mast Cells/chemistry , Rats , Staining and Labeling
20.
Molecules ; 20(3): 4254-76, 2015 Mar 05.
Article En | MEDLINE | ID: mdl-25751786

Heparan sulfate (HS) and heparin are glycosaminoglycans (GAGs) that are heterogeneous in nature, not only due to differing disaccharide combinations, but also their sulfate modifications. HS is well known for its interactions with various growth factors and cytokines; and heparin for its clinical use as an anticoagulant. Due to their potential use in tissue regeneration; and the recent adverse events due to contamination of heparin; there is an increased surge to produce these GAGs on a commercial scale. The production of HS from natural sources is limited so strategies are being explored to be biomimetically produced via chemical; chemoenzymatic synthesis methods and through the recombinant expression of proteoglycans. This review details the most recent advances in the field of HS/heparin synthesis for the production of low molecular weight heparin (LMWH) and as a tool further our understanding of the interactions that occur between GAGs and growth factors and cytokines involved in tissue development and repair.


Anticoagulants/metabolism , Biomimetics , Heparin/metabolism , Heparitin Sulfate/metabolism , Animals , Anticoagulants/chemistry , Heparin/chemistry , Heparitin Sulfate/chemistry , Humans
...