51
|
Wójcik-Pastuszka D, Skrzypczyk A, Musiał W. The Interactions and Release Kinetics of Sodium Hyaluronate Implemented in Nonionic and Anionic Polymeric Hydrogels, Studied by Immunoenzymatic ELISA Test. Pharmaceutics 2021; 14:58. [PMID: 35056954 PMCID: PMC8779175 DOI: 10.3390/pharmaceutics14010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Hyaluronan is a natural polymer that was introduced to wound therapy. Formulations based on synthetic polymers such as methylcellulose (MC) and polyacrylic acid (PA) containing hyaluronan (HA) were proposed for the development of prospective wound-healing preparations. The formulations of different carrier concentrations containing a fixed amount of HA were prepared, and their viscosity was measured. The HA release was evaluated by employing the apparatus paddle over a disc. The hydrogels were introduced to the donor chamber, and HA was released to the pH = 7.4 buffer. The amount of HA released was obtained using the ELISA test. The release was analyzed on the basis of different kinetic models: zero-, first-, and second-order kinetics, as well as Higuchi and Korsmeyer-Peppas equations. The release rate constants and the half release time were calculated from these equations. According to the value of the coefficient of the determination, the best model describing the observed process was selected. The comparison between the dissolution profiles was carried out by calculating the difference factor f1 and the similarity factor f2. The interaction between the hydrogel components was investigated by Fourier-transform infrared spectroscopy (FTIR) and differential scanning calorimetry (DSC) measurements. The study revealed that the zero-order equation best described the release of HA from the formulations studied. The FTIR research and the DSC study showed the intermolecular interaction between HA chains in MC-based compositions, as well as between HA and the synthetic polymer in the PA-based formulations. The study revealed that the formulation with a higher concentration of synthetic polymer may prolong the release of HA. The obtained results indicated that the proposed hydrogels have potential for wound healing and may accelerate skin regeneration.
Collapse
Affiliation(s)
| | | | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211A, 55-556 Wroclaw, Poland; (D.W.-P.); (A.S.)
| |
Collapse
|
52
|
Lazarus E, Bermudez-Lekerika P, Farchione D, Schofield T, Howard S, Mambetkadyrov I, Lamoca M, Rivero IV, Gantenbein B, Lewis CL, Wuertz-Kozak K. Sulfated Hydrogels in Intervertebral Disc and Cartilage Research. Cells 2021; 10:cells10123568. [PMID: 34944076 PMCID: PMC8700363 DOI: 10.3390/cells10123568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 01/07/2023] Open
Abstract
Hydrogels are commonly used for the 3D culture of musculoskeletal cells. Sulfated hydrogels, which have seen a growing interest over the past years, provide a microenvironment that help maintain the phenotype of chondrocytes and chondrocyte-like cells and can be used for sustained delivery of growth factors and other drugs. Sulfated hydrogels are hence valuable tools to improve cartilage and intervertebral disc tissue engineering. To further advance the utilization of these hydrogels, we identify and summarize the current knowledge about different sulfated hydrogels, highlight their beneficial effects in cartilage and disc research, and review the biofabrication processes most suitable to secure best quality assurance through deposition fidelity, repeatability, and attainment of biocompatible morphologies.
Collapse
Affiliation(s)
- Emily Lazarus
- Department of Industrial and Systems Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (E.L.); (I.V.R.)
| | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
| | - Daniel Farchione
- Inamori School of Engineering, Alfred University, Alfred, NY 14802, USA;
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Taylor Schofield
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Sloan Howard
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Iskender Mambetkadyrov
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Mikkael Lamoca
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Iris V. Rivero
- Department of Industrial and Systems Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (E.L.); (I.V.R.)
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
| | - Christopher L. Lewis
- Department of Manufacturing and Mechanical Engineering Technology, Rochester Institute of Technology, Rochester, NY 14632, USA;
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
- Schoen Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (AU), 81547 Munich, Germany
- Correspondence: ; Tel.: +1-585-475-7355
| |
Collapse
|
53
|
Lu YH, Baker AEG, Fokina A, Kufleitner M, Kivijäri T, Shoichet MS. Temporally Controlled Photouncaged Epidermal Growth Factor Influences Cell Fate in Hydrogels. ACS Biomater Sci Eng 2021; 8:185-195. [PMID: 34860498 DOI: 10.1021/acsbiomaterials.1c00941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hydrogels are powerful materials that more accurately mimic the cellular microenvironment over static two-dimensional culture. Photochemical strategies enable dynamic complexity to be achieved within hydrogels to better mimic the extracellular matrix; however, many photochemical systems to pattern proteins within hydrogels are complicated by long reaction times to immobilize these proteins wherein the protein can lose activity. As proof-of-concept, we demonstrate an elegant method where photocaged proteins are immobilized in hydrogels and then directly photoactivated. Specifically, we immobilized streptavidin-ortho-nitrobenzyl-modified epidermal growth factor (EGF) to cross-linked hyaluronan hydrogels and cultured two EGF-responsive cancer cells of breast and lung therein. We used light to temporally uncage and control EGF activation, thereby inducing cell death in breast cancer cells and proliferation in lung cancer cells. These results show how temporal, photochemical, protein activation influences cellular response and lays the foundation for further advances in manipulating the in vitro environment to control cell fate.
Collapse
Affiliation(s)
- Yung Hsiang Lu
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Alexander E G Baker
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Ana Fokina
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Markus Kufleitner
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada.,Department of Chemistry, Universität Konstanz, D-78457 Konstanz, Germany
| | - Tove Kivijäri
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada.,Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden
| | - Molly S Shoichet
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
54
|
Hauck S, Zager P, Halfter N, Wandel E, Torregrossa M, Kakpenova A, Rother S, Ordieres M, Räthel S, Berg A, Möller S, Schnabelrauch M, Simon JC, Hintze V, Franz S. Collagen/hyaluronan based hydrogels releasing sulfated hyaluronan improve dermal wound healing in diabetic mice via reducing inflammatory macrophage activity. Bioact Mater 2021; 6:4342-4359. [PMID: 33997511 PMCID: PMC8105600 DOI: 10.1016/j.bioactmat.2021.04.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Sustained inflammation associated with dysregulated macrophage activation prevents tissue formation and healing of chronic wounds. Control of inflammation and immune cell functions thus represents a promising approach in the development of advanced therapeutic strategies. Here we describe immunomodulatory hyaluronan/collagen (HA-AC/coll)-based hydrogels containing high-sulfated hyaluronan (sHA) as immunoregulatory component for the modulation of inflammatory macrophage activities in disturbed wound healing. Solute sHA downregulates inflammatory activities of bone marrow-derived and tissue-resident macrophages in vitro. This further affects macrophage-mediated pro-inflammatory activation of skin cells as shown in skin ex-vivo cultures. In a mouse model of acute skin inflammation, intradermal injection of sHA downregulates the inflammatory processes in the skin. This is associated with the promotion of an anti-inflammatory gene signature in skin macrophages indicating a shift of their activation profile. For in vivo translation, we designed HA-AC/coll hydrogels allowing delivery of sHA into wounds over a period of at least one week. Their immunoregulatory capacity was analyzed in a translational experimental approach in skin wounds of diabetic db/db mice, an established model for disturbed wound healing. The sHA-releasing hydrogels improved defective tissue repair with reduced inflammation, augmented pro-regenerative macrophage activation, increased vascularization, and accelerated new tissue formation and wound closure.
Collapse
Affiliation(s)
- Sophia Hauck
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Paula Zager
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Norbert Halfter
- Institute of Materials Science, Max Bergmann Center for Biomaterials, Technische Universität Dresden, 01069, Dresden, Germany
| | - Elke Wandel
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Marta Torregrossa
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Ainur Kakpenova
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center for Biomaterials, Technische Universität Dresden, 01069, Dresden, Germany
| | - Michelle Ordieres
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Susann Räthel
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Albrecht Berg
- Biomaterials Department, INNOVENT e.V. Jena, Germany
| | | | | | - Jan C. Simon
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center for Biomaterials, Technische Universität Dresden, 01069, Dresden, Germany
| | - Sandra Franz
- Department of Dermatology, Venerology und Allergology, Leipzig University, 04103, Leipzig, Germany
- Corresponding author. University Leipzig, Department of Dermatology, Venerology and Allergology, Max Bürger Research Centre, Johannisallee 30, 04103, Leipzig, Germany.
| |
Collapse
|
55
|
Balamurugan K, Pisabarro MT. Stabilizing Role of Water Solvation on Anion-π Interactions in Proteins. ACS OMEGA 2021; 6:25350-25360. [PMID: 34632193 PMCID: PMC8495695 DOI: 10.1021/acsomega.1c03264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/20/2021] [Indexed: 05/31/2023]
Abstract
In this work, anion-π interactions between sulfate groups (SO4 2-) and protein aromatic amino acids (AAs) (histidine protonated (HisP), histidine neutral (HisN), tyrosine (Tyr), tryptophan (Trp), and phenylalanine (Phe)) in an aqueous environment have been analyzed using quantum chemical (QC) calculations and molecular dynamics (MD) simulations. Sulfates can occur naturally in solution and can be contained in biomolecules playing relevant roles in their biological function. In particular, the presence of sulfate groups in glycosaminoglycans such as heparin and heparan sulfate has been shown to be relevant for protein and cellular communication and, consequently, for tissue regeneration. Therefore, anion-π interactions between sulfate groups and aromatic residues represent a relevant aspect to investigate. QC results show that such an anion-π mode of interaction between SO4 2- and aromatic AAs is only possible in the presence of water molecules, in the absence of any other cooperative non-covalent interactions. Protonated histidine stands out in terms of its enhancement in the magnitude of interaction strength on solvation. Other AAs such as non-protonated histidine, tyrosine, and phenylalanine can stabilize anion-π interactions on solvation, albeit with weak interaction energy. Tryptophan does not exhibit any anion-π mode of interaction with SO4 2-. The order of magnitude of the interaction of aromatic AAs with SO4 2- on microsolvation is HisP > HisN > Tyr > Trp > Phe. Atoms in molecules (AIM) analysis illustrates the significance of water molecules in stabilizing the divalent SO4 2- anion over the π surface of the aromatic AAs. MD simulation analysis shows that the order of magnitude of the interaction of SO4 2- with aromatic AAs in macroscopic solvation is HisP > HisN, Tyr, Trp > Phe, which is very much in line with the QC results. Spatial distribution function analysis illustrates that protonated histidine alone is capable of establishing the anion-π interaction with SO4 2- in the solution phase. This study sheds light on the understanding of anion-π interactions between SO4 2- and aromatic AAs such as His and Tyr observed in protein crystal structures and the significance of water molecules in stabilizing such interactions, which is not feasible otherwise.
Collapse
|
56
|
Spiller S, Wippold T, Bellmann-Sickert K, Franz S, Saalbach A, Anderegg U, Beck-Sickinger AG. Protease-Triggered Release of Stabilized CXCL12 from Coated Scaffolds in an Ex Vivo Wound Model. Pharmaceutics 2021; 13:pharmaceutics13101597. [PMID: 34683890 PMCID: PMC8539926 DOI: 10.3390/pharmaceutics13101597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Biomaterials are designed to improve impaired healing of injured tissue. To accomplish better cell integration, we suggest to coat biomaterial surfaces with bio-functional proteins. Here, a mussel-derived surface-binding peptide is used and coupled to CXCL12 (stromal cell-derived factor 1α), a chemokine that activates CXCR4 and consequently recruits tissue-specific stem and progenitor cells. CXCL12 variants with either non-releasable or protease-mediated-release properties were designed and compared. Whereas CXCL12 was stabilized at the N-terminus for protease resistance, a C-terminal linker was designed that allowed for specific cleavage-mediated release by matrix metalloproteinase 9 and 2, since both enzymes are frequently found in wound fluid. These surface adhesive CXCL12 derivatives were produced by expressed protein ligation. Functionality of the modified chemokines was assessed by inositol phosphate accumulation and cell migration assays. Increased migration of keratinocytes and primary mesenchymal stem cells was demonstrated. Immobilization and release were studied for bioresorbable PCL-co-LC scaffolds, and accelerated wound closure was demonstrated in an ex vivo wound healing assay on porcine skin grafts. After 24 h, a significantly improved CXCL12-specific growth stimulation of the epithelial tips was already observed. The presented data display a successful application of protein-coated biomaterials for skin regeneration.
Collapse
Affiliation(s)
- Sabrina Spiller
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
| | - Tom Wippold
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
| | - Sandra Franz
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Anja Saalbach
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
- Correspondence: (U.A.); (A.G.B.-S.); Tel.: +49-341-972-5881 (U.A.); +49-341-973-6900 (A.G.B.-S.); Fax: +49-341-972-5878 (U.A.); +49-341-973-6909 (A.G.B.-S.)
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
- Correspondence: (U.A.); (A.G.B.-S.); Tel.: +49-341-972-5881 (U.A.); +49-341-973-6900 (A.G.B.-S.); Fax: +49-341-972-5878 (U.A.); +49-341-973-6909 (A.G.B.-S.)
| |
Collapse
|
57
|
Superior Technique for the Production of Agarose Dressing Containing Sericin and Its Wound Healing Property. Polymers (Basel) 2021; 13:polym13193370. [PMID: 34641182 PMCID: PMC8512865 DOI: 10.3390/polym13193370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 12/28/2022] Open
Abstract
Finding a simple and eco-friendly production technique that matches to the natural agent and results in a truly valuable natural scaffold production is still limited amongst the intensively competitive natural scaffold development. Therefore, the purpose of this study was to develop natural scaffolds that were environmentally friendly, low cost, and easily produced, using natural agents and a physical crosslinking technique. These scaffolds were prepared from agarose and sericin using the freeze-drying method (D) or freeze-thawing together with the freeze-drying method (TD). Moreover, plasticizers were added into the scaffold to improve their properties. Their physical, mechanical, and biological properties were investigated. The results showed that scaffolds that were prepared using the TD method had stronger bonding between sericin and other compounds, leading to a low swelling ratio and low protein release of the scaffolds. This property may be applied in the development of further material as a controlled drug release scaffold. Adding plasticizers, especially glycerin, into the scaffolds significantly increased elongation properties, leading to an increase in elasticity of the scaffold. Moreover, all scaffolds could activate cell migration, which had an advantage on wound healing acceleration. Accordingly, this study was successful in developing natural scaffolds using natural agents and simple and green crosslinking methods.
Collapse
|
58
|
Otsuka T, Kan HM, Laurencin CT. Regenerative Engineering Approaches to Scar-Free Skin Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021. [DOI: 10.1007/s40883-021-00229-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
59
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
60
|
Rial-Hermida MI, Rey-Rico A, Blanco-Fernandez B, Carballo-Pedrares N, Byrne EM, Mano JF. Recent Progress on Polysaccharide-Based Hydrogels for Controlled Delivery of Therapeutic Biomolecules. ACS Biomater Sci Eng 2021; 7:4102-4127. [PMID: 34137581 PMCID: PMC8919265 DOI: 10.1021/acsbiomaterials.0c01784] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 06/02/2021] [Indexed: 12/24/2022]
Abstract
A plethora of applications using polysaccharides have been developed in recent years due to their availability as well as their frequent nontoxicity and biodegradability. These polymers are usually obtained from renewable sources or are byproducts of industrial processes, thus, their use is collaborative in waste management and shows promise for an enhanced sustainable circular economy. Regarding the development of novel delivery systems for biotherapeutics, the potential of polysaccharides is attractive for the previously mentioned properties and also for the possibility of chemical modification of their structures, their ability to form matrixes of diverse architectures and mechanical properties, as well as for their ability to maintain bioactivity following incorporation of the biomolecules into the matrix. Biotherapeutics, such as proteins, growth factors, gene vectors, enzymes, hormones, DNA/RNA, and antibodies are currently in use as major therapeutics in a wide range of pathologies. In the present review, we summarize recent progress in the development of polysaccharide-based hydrogels of diverse nature, alone or in combination with other polymers or drug delivery systems, which have been implemented in the delivery of biotherapeutics in the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- M. Isabel Rial-Hermida
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| | - Ana Rey-Rico
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Barbara Blanco-Fernandez
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology, 08028 Barcelona, Spain
- CIBER
en Bioingeniería, Biomateriales y
Nanomedicina, CIBER-BBN, 28029 Madrid, Spain
| | - Natalia Carballo-Pedrares
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Eimear M. Byrne
- Wellcome-Wolfson
Institute For Experimental Medicine, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - João F. Mano
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| |
Collapse
|
61
|
Miao F, Li Y, Tai Z, Zhang Y, Gao Y, Hu M, Zhu Q. Antimicrobial Peptides: The Promising Therapeutics for Cutaneous Wound Healing. Macromol Biosci 2021; 21:e2100103. [PMID: 34405955 DOI: 10.1002/mabi.202100103] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/31/2021] [Indexed: 12/12/2022]
Abstract
Chronic wound infections have caused an increasing number of deaths and economic burden, which necessitates wound treatment options. Hitherto, the development of functional wound dressings has achieved reasonable progress. Antibacterial agents, growth factors, and miRNAs are incorporated in different wound dressings to treat various types of wounds. As an effective antimicrobial agent and emerging wound healing therapeutic, antimicrobial peptides (AMPs) have attracted significant attention. The present study focuses on the application of AMPs in wound healing and discusses the types, properties and formulation strategies of AMPs used for wound healing. In addition, the clinical trial and the current status of studies on "antimicrobial peptides and wound healing" are elaborated through bibliometrics. Also, the challenges and opportunities for further development and utilization of AMP formulations in wound healing are discussed.
Collapse
Affiliation(s)
- Fengze Miao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Ying Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China
| | - Yong Zhang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Yue Gao
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Menghong Hu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China
| |
Collapse
|
62
|
Spiller S, Clauder F, Bellmann-Sickert K, Beck-Sickinger AG. Improvement of wound healing by the development of ECM-inspired biomaterial coatings and controlled protein release. Biol Chem 2021; 402:1271-1288. [PMID: 34392636 DOI: 10.1515/hsz-2021-0144] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/19/2021] [Indexed: 12/22/2022]
Abstract
Implant design has evolved from biochemically inert substrates, minimizing cell and protein interaction, towards sophisticated bioactive substrates, modulating the host response and supporting the regeneration of the injured tissue. Important aspects to consider are the control of cell adhesion, the discrimination of bacteria and non-local cells from the desired tissue cell type, and the stimulation of implant integration and wound healing. Here, the extracellular matrix acts as a role model providing us with inspiration for sophisticated designs. Within this scope, small bioactive peptides have proven to be miscellaneously deployable for the mediation of surface, cell and matrix interactions. Combinations of adhesion ligands, proteoglycans, and modulatory proteins should guide multiple aspects of the regeneration process and cooperativity between the different extracellular matrix components, which bears the chance to maximize the therapeutic efficiency and simultaneously lower the doses. Hence, efforts to include multiple of these factors in biomaterial design are well worth. In the following, multifunctional implant coatings based on bioactive peptides are reviewed and concepts to implement strong surface anchoring for stable cell adhesion and a dynamic delivery of modulator proteins are discussed.
Collapse
Affiliation(s)
- Sabrina Spiller
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| |
Collapse
|
63
|
Heydari P, Kharaziha M, Varshosaz J, Javanmard SH. Current knowledge of immunomodulation strategies for chronic skin wound repair. J Biomed Mater Res B Appl Biomater 2021; 110:265-288. [PMID: 34318595 DOI: 10.1002/jbm.b.34921] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/11/2022]
Abstract
In orchestrating the wound healing process, the immune system plays a critical role. Hence, controlling the immune system to repair skin defects is an attractive approach. The highly complex immune system includes the coordinated actions of several immune cells, which can produce various inflammatory and antiinflammatory cytokines and affect the healing of skin wounds. This process can be optimized using biomaterials, bioactive molecules, and cell delivery. The present review discusses various immunomodulation strategies for supporting the healing of chronic wounds. In this regard, following the evolution of the immune system and its role in the wound healing mechanism, the interaction between the extracellular mechanism and immune cells for acceleration wound healing will be firstly investigated. Consequently, the immune-based chronic wounds will be briefly examined and the mechanism of progression, and conventional methods of their treatment are evaluated. In the following, various biomaterials-based immunomodulation strategies are introduced to stimulate and control the immune system to treat and regenerate skin defects. Other effective methods of controlling the immune system in wound healing which is the release of bioactive agents (such as antiinflammatory, antigens, and immunomodulators) and stem cell therapy at the site of injury are reviewed.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Jaleh Varshosaz
- School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
64
|
Zhao X, Li S, Ding J, Wei J, Tian P, Wei H, Chen T. Combination of an engineered Lactococcus lactis expressing CXCL12 with light-emitting diode yellow light as a treatment for scalded skin in mice. Microb Biotechnol 2021; 14:2090-2100. [PMID: 34310856 PMCID: PMC8449663 DOI: 10.1111/1751-7915.13885] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/14/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
Impaired wound closure is an increasingly crucial clinical challenge. Recently, wound healing has shifted towards innovative treatments that exploit nanotechnology, biomaterials, biologics and phototherapy. Here, we constructed an engineered MG1363-pMG36e-mCXCL12 strain with pMG36e plasmid encoding stromal cell-derived factor 1α (named CXCL12) and evaluated the synergistic effects of light-emitting diode (LED) yellow light and MG1363-pMG36e-mCXCL12 on scald wounds in mice. The results indicated that the combined treatment with LED yellow light with mCXCL12 delivering strain accelerated wound closure, tissue remodelling, re-epithelialization and hair follicle regeneration and inhibited over-inflammation oppositely in the central and surrounding wounds by macroscopic, histopathologic and immunohistochemistry parameters. Furthermore, combination therapy increased the epidermal growth factor and Ki67-positive cells and upregulated beta-catenin (β-catenin), cellular-myelocytomatosis (c-Myc), wingless-type MMTV integration site family member 1 (Wnt1), Jagged 1, neurogenic locus notch homolog protein 1 (Notch 1) and hairy and enhancer of split 1 (Hes 1) protein levels of the Wnt and Notch signalling pathways. It also facilitated collagen fibrogenesis and deposition and improved the activities of hydroxyproline, superoxide dismutase and glutathione peroxidase in scalded granulation tissue, in addition to reducing the inflammatory factors interleukin 1 beta (IL-1β) and tumour necrosis factor alpha (TNF-α). The combined treatment effectively reduced skin pathogens Ralstonia and Acinetobacter to further reduce the risk of infection. Overall, combination of LED yellow light and MG1363-pMG36e-mCXCL12 represents a potential strategy for the treatment of cutaneous wounds.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Shengjie Li
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Jianing Ding
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Jing Wei
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Puyuan Tian
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Hong Wei
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Tingtao Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
| |
Collapse
|
65
|
Schiller J, Lemmnitzer K, Dürig JN, Rademann J. Insights into structure, affinity, specificity, and function of GAG-protein interactions through the chemoenzymatic preparation of defined sulfated oligohyaluronans. Biol Chem 2021; 402:1375-1384. [PMID: 34291624 DOI: 10.1515/hsz-2021-0165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/04/2021] [Indexed: 11/15/2022]
Abstract
High amounts of glycosaminoglycans (GAG) such as hyaluronan (HA) occur in connective tissues. There is nowadays increasing evidence that a "sulfation code" exists which mediates numerous GAG functions. High molecular weight and inhomogeneity of GAG, however, aggravated detailed studies. Thus, synthetic oligosaccharides were urgently required. We will review here chemoenzymatic and analytic strategies to provide defined sulfated and anomerically modified GAG oligosaccharides of the HA type. Representative studies of protein/GAG interactions by (bio)chemical and biophysical methods are reported yielding novel insights into GAG-protein binding. Finally, the biological conclusions and in vivo applications of defined sulfated GAG oligosaccharides will be discussed.
Collapse
Affiliation(s)
- Jürgen Schiller
- Faculty of Medicine, Institute of Medical Physics and Biophysics, University of Leipzig, Härtelstraße 16-18, 04107Leipzig, Germany
| | - Katharina Lemmnitzer
- Faculty of Medicine, Institute of Medical Physics and Biophysics, University of Leipzig, Härtelstraße 16-18, 04107Leipzig, Germany
| | - Jan-Niklas Dürig
- Department of Biology, Chemistry, and Pharmacy, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195Berlin, Germany
| | - Jörg Rademann
- Department of Biology, Chemistry, and Pharmacy, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195Berlin, Germany
| |
Collapse
|
66
|
Balamurugan K, Koehler L, Dürig JN, Hempel U, Rademann J, Hintze V, Pisabarro MT. Structural insights into the modulation of PDGF/PDGFR-β complexation by hyaluronan derivatives. Biol Chem 2021; 402:1441-1452. [PMID: 34280958 DOI: 10.1515/hsz-2021-0173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022]
Abstract
Angiogenesis is an important physiological process playing a crucial role in wound healing and cancer progression. Vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) are key players in angiogenesis. Based on previous findings regarding the modulation of VEGF activity by glycosaminoglycans (GAG), here we explore the interaction of hyaluronan (HA)-based GAG with PDGF and its receptor PDGFR-β by applying molecular modeling and dynamics simulations in combination with surface plasmon resonance (SPR). Computational analysis on the interaction of oligo-hyaluronan derivatives with different sulfation pattern and functionalization shows that these GAG interact with PDGF in relevant regions for receptor recognition, and that high sulfation as well as modification with the TAMRA group convey stronger binding. On the other hand, the studied oligo-hyaluronan derivatives are predicted to scarcely recognize PDGFR-β. SPR results are in line with the computational predictions regarding the binding pattern of HA tetrasaccharide (HA4) derivatives to PDGF and PDGFR-β. Furthermore, our experimental results also show that the complexation of PDGF to PDGFR-β can be modulated by HA4 derivatives. The results found open the path for considering HA4 derivatives as potential candidates to be exploited for modulation of the PDGF/PDGFR-β signaling system in angiogenesis and related disease conditions.
Collapse
Affiliation(s)
- Kanagasabai Balamurugan
- Structural Bioinformatics, BIOTEC Technische Universität Dresden, Tatzberg 47-51, D-01307Dresden, Germany
| | - Linda Koehler
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069Dresden, Germany
| | - Jan-Niklas Dürig
- Medicinal Chemistry Department, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, D-14195Berlin, Germany
| | - Ute Hempel
- Institute of Physiological Chemistry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fiedlerstraße 42, D-01307Dresden, Germany
| | - Jörg Rademann
- Medicinal Chemistry Department, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, D-14195Berlin, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069Dresden, Germany
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC Technische Universität Dresden, Tatzberg 47-51, D-01307Dresden, Germany
| |
Collapse
|
67
|
Ceylan S. An in vitro evaluation of Genipin‐crosslinked and
Hypericum perforatum
incorporated novel membranes for skin tissue engineering applications. J Appl Polym Sci 2021. [DOI: 10.1002/app.51385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Seda Ceylan
- Bioengineering Department, Faculty of Engineering Adana Alparslan Türkeş Science and Technology University Adana Turkey
| |
Collapse
|
68
|
Anderegg U, Halfter N, Schnabelrauch M, Hintze V. Collagen/glycosaminoglycan-based matrices for controlling skin cell responses. Biol Chem 2021; 402:1325-1335. [PMID: 34218546 DOI: 10.1515/hsz-2021-0176] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022]
Abstract
Wound healing and tissue regeneration are orchestrated by the cellular microenvironment, e.g. the extracellular matrix (ECM). Including ECM components in biomaterials is a promising approach for improving regenerative processes, e.g. wound healing in skin. This review addresses recent findings for enhanced epidermal-dermal regenerative processes on collagen (coll)/glycosaminoglycan (GAG)-based matrices containing sulfated GAG (sGAG) in simple and complex in vitro models. These matrices comprise 2D-coatings, electrospun nanofibrous scaffolds, and photo-crosslinked acrylated hyaluronan (HA-AC)/coll-based hydrogels. They demonstrated to regulate keratinocyte and fibroblast migration and growth, to stimulate melanogenesis in melanocytes from the outer root sheath (ORS) of hair follicles and to enhance the epithelial differentiation of human mesenchymal stem cells (hMSC). The matrices' suitability for delivery of relevant growth factors, like heparin-binding epidermal growth factor like growth factor (HB-EGF), further highlights their potential as bioinspired, functional microenvironments for enhancing skin regeneration.
Collapse
Affiliation(s)
- Ulf Anderegg
- Department of Dermatology, Venereology and Allergology, Leipzig University, D-04103Leipzig, Germany
| | - Norbert Halfter
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069 Dresden, Germany
| | | | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069 Dresden, Germany
| |
Collapse
|
69
|
Kühn S, Freyse J, Atallah P, Rademann J, Freudenberg U, Werner C. Tuning the network charge of biohybrid hydrogel matrices to modulate the release of SDF-1. Biol Chem 2021; 402:1453-1464. [PMID: 34218538 DOI: 10.1515/hsz-2021-0175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/10/2021] [Indexed: 11/15/2022]
Abstract
The delivery of chemotactic signaling molecules via customized biomaterials can effectively guide the migration of cells to improve the regeneration of damaged or diseased tissues. Here, we present a novel biohybrid hydrogel system containing two different sulfated glycosaminoglycans (sGAG)/sGAG derivatives, namely either a mixture of short heparin polymers (Hep-Mal) or structurally defined nona-sulfated tetrahyaluronans (9s-HA4-SH), to precisely control the release of charged signaling molecules. The polymer networks are described in terms of their negative charge, i.e. the anionic sulfate groups on the saccharides, using two parameters, the integral density of negative charge and the local charge distribution (clustering) within the network. The modulation of both parameters was shown to govern the release characteristics of the chemotactic signaling molecule SDF-1 and allows for seamless transitions between burst and sustained release conditions as well as the precise control over the total amount of delivered protein. The obtained hydrogels with well-adjusted release profiles effectively promote MSC migration in vitro and emerge as promising candidates for new treatment modalities in the context of bone repair and wound healing.
Collapse
Affiliation(s)
- Sebastian Kühn
- Leibniz Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Str. 6, D-01069Dresden, Germany
| | - Joanna Freyse
- Institute of Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Strasse 2+4, D-14195Berlin, Germany
| | - Passant Atallah
- Leibniz Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Str. 6, D-01069Dresden, Germany
| | - Jörg Rademann
- Institute of Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Strasse 2+4, D-14195Berlin, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Str. 6, D-01069Dresden, Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Str. 6, D-01069Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstraße 105, D-01307Dresden, Germany
| |
Collapse
|
70
|
Engineering of cerium oxide loaded chitosan/polycaprolactone hydrogels for wound healing management in model of cardiovascular surgery. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.03.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
71
|
Ali AF, Ahmed MM, El-Kady AM, Abd El-Hady BM, Ibrahim AM. Synthesis of Gelatin-Agarose Scaffold for Controlled Antibiotic Delivery and its Modification by Glass Nanoparticles Addition as a Potential Osteomyelitis Treatment. SILICON 2021; 13:2011-2028. [DOI: 10.1007/s12633-020-00576-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 06/26/2020] [Indexed: 09/02/2023]
|
72
|
Schnabelrauch M, Schiller J, Möller S, Scharnweber D, Hintze V. Chemically modified glycosaminoglycan derivatives as building blocks for biomaterial coatings and hydrogels. Biol Chem 2021; 402:1385-1395. [PMID: 34008374 DOI: 10.1515/hsz-2021-0171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/07/2021] [Indexed: 12/21/2022]
Abstract
Tissue regeneration is regulated by the cellular microenvironment, e.g. the extracellular matrix. Here, sulfated glycosaminoglycans (GAG), are of vital importance interacting with mediator proteins and influencing their biological activity. Hence, they are promising candidates for controlling tissue regeneration. This review addresses recent achievements regarding chemically modified GAG as well as collagen/GAG-based coatings and hydrogels including (i) chemical functionalization strategies for native GAG, (ii) GAG-based biomaterial strategies for controlling cellular responses, (iii) (bio)chemical methods for characterization and iv) protein interaction profiles and attained tissue regeneration in vitro and in vivo. The potential of GAG for bioinspired, functional biomaterials is highlighted.
Collapse
Affiliation(s)
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, Medical Faculty, Universität Leipzig, D-04107 Leipzig, Germany
| | - Stephanie Möller
- Biomaterials Department, INNOVENT e.V., Prüssingstrasse 27B, D-07745Jena, Germany
| | - Dieter Scharnweber
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, D-01069Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, D-01069Dresden, Germany
| |
Collapse
|
73
|
Sustained Release Systems for Delivery of Therapeutic Peptide/Protein. Biomacromolecules 2021; 22:2299-2324. [PMID: 33957752 DOI: 10.1021/acs.biomac.1c00160] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peptide/protein therapeutics have been significantly applied in the clinical treatment of various diseases such as cancer, diabetes, etc. owing to their high biocompatibility, specificity, and therapeutic efficacy. However, due to their immunogenicity, instability stemming from its complex tertiary and quaternary structure, vulnerability to enzyme degradation, and rapid renal clearance, the clinical application of protein/peptide therapeutics is significantly confined. Though nanotechnology has been demonstrated to prevent enzyme degradation of the protein therapeutics and thus enhance the half-life, issues such as initial burst release and uncontrollable release kinetics are still unsolved. Moreover, the traditional administration method results in poor patient compliance, limiting the clinical application of protein/peptide therapeutics. Exploiting the sustained-release formulations for more controllable delivery of protein/peptide therapeutics to decrease the frequency of injection and enhance patient compliance is thus greatly meaningful. In this review, we comprehensively summarize the substantial advancements of protein/peptide sustained-release systems in the past decades. In addition, the advantages and disadvantages of all these sustained-release systems in clinical application together with their future challenges are also discussed in this review.
Collapse
|
74
|
Souza PR, de Oliveira AC, Vilsinski BH, Kipper MJ, Martins AF. Polysaccharide-Based Materials Created by Physical Processes: From Preparation to Biomedical Applications. Pharmaceutics 2021; 13:621. [PMID: 33925380 PMCID: PMC8146878 DOI: 10.3390/pharmaceutics13050621] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Polysaccharide-based materials created by physical processes have received considerable attention for biomedical applications. These structures are often made by associating charged polyelectrolytes in aqueous solutions, avoiding toxic chemistries (crosslinking agents). We review the principal polysaccharides (glycosaminoglycans, marine polysaccharides, and derivatives) containing ionizable groups in their structures and cellulose (neutral polysaccharide). Physical materials with high stability in aqueous media can be developed depending on the selected strategy. We review strategies, including coacervation, ionotropic gelation, electrospinning, layer-by-layer coating, gelation of polymer blends, solvent evaporation, and freezing-thawing methods, that create polysaccharide-based assemblies via in situ (one-step) methods for biomedical applications. We focus on materials used for growth factor (GFs) delivery, scaffolds, antimicrobial coatings, and wound dressings.
Collapse
Affiliation(s)
- Paulo R. Souza
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
| | - Ariel C. de Oliveira
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
- Laboratory of Materials, Macromolecules and Composites, Federal University of Technology—Paraná (UTFPR), Apucarana 86812-460, PR, Brazil
| | - Bruno H. Vilsinski
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
| | - Matt J. Kipper
- Department of Chemical and Biological Engineering, Colorado State University (CSU), Fort Collins, CO 80523, USA
- School of Advanced Materials Discovery, Colorado State University (CSU), Fort Collins, CO 80523, USA
- School of Biomedical Engineering, Colorado State University (CSU), Fort Collins, CO 80523, USA
| | - Alessandro F. Martins
- Group of Polymeric Materials and Composites, Department of Chemistry, State University of Maringá (UEM), Maringá 87020-900, PR, Brazil; (P.R.S.); (A.C.d.O.); (B.H.V.)
- Laboratory of Materials, Macromolecules and Composites, Federal University of Technology—Paraná (UTFPR), Apucarana 86812-460, PR, Brazil
- Department of Chemical and Biological Engineering, Colorado State University (CSU), Fort Collins, CO 80523, USA
| |
Collapse
|
75
|
Grudén S, Brunmark C, Holmqvist B, Brenndörfer ED, Johansson M, Liu J, Zhao Y, Axén N, Hassan M. Biodistribution of fluorescence-labelled EGF protein from slow release NanoZolid depots in mouse. Int J Pharm 2021; 601:120588. [PMID: 33845148 DOI: 10.1016/j.ijpharm.2021.120588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 03/05/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
AIM The study was designed to evaluate the ability of the calcium sulfate based NanoZolid® drug delivery technology to locally release the epidermal growth factor (EGF) protein while maintaining its biological activity. METHODS NanoZolid-formulated EGF protein labelled with a near infrared dye (EGF-NIR) depots or EGF-NIR dissolved in PBS were injected subcutaneously into mice bearing EGF receptor (EGFR) positive human A549 lung cancer tumors inoculated subcutaneously. The release and biodistribution of the EGF-NIR were investigated in vivo longitudinally up to 96 h post administration, utilizing whole body fluorescence imaging. In order to confirm the in vivo findings, histological analysis of tumor cryosections was performed to investigate EGF-NIR fluorescent signal and EGFR expression level by immunofluorescence labelling. RESULTS The in vivo fluorescence imaging showed a controlled release profile of the EGF-NIR loaded in the NanoZolid depots compared to free EGF-NIR. Histological analysis of the tumors further demonstrated a prevailing distribution of EGF-NIR in regions with high levels of EGFR expression. CONCLUSION Calcium sulfate based depots can be used to formulate EGF while maintaining its biological activity, e.g. receptor binding capability. This may have a good clinical potential for local delivery of biomolecules to enhance treatment efficacy and minimize systemic adverse effects.
Collapse
Affiliation(s)
- Stefan Grudén
- Experimental Cancer Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden; LIDDS AB, Virdings allé 32B, SE-754 50 Uppsala, Sweden.
| | - Charlott Brunmark
- Truly Labs AB, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden.
| | - Bo Holmqvist
- ImaGene-iT AB, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden.
| | | | | | - Jian Liu
- Truly Labs AB, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden.
| | - Ying Zhao
- Experimental Cancer Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden; Clinical Research Center (KFC) and Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | - Niklas Axén
- LIDDS AB, Virdings allé 32B, SE-754 50 Uppsala, Sweden.
| | - Moustapha Hassan
- Experimental Cancer Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden; Clinical Research Center (KFC) and Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
76
|
Abune L, Wang Y. Affinity Hydrogels for Protein Delivery. Trends Pharmacol Sci 2021; 42:300-312. [PMID: 33632537 PMCID: PMC7954985 DOI: 10.1016/j.tips.2021.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/24/2022]
Abstract
Proteins have been studied as therapeutic agents for treatment of various human diseases. However, the delivery of protein drugs into the body is challenging. In this review, we summarize and highlight progress in developing affinity hydrogels (i.e., hydrogels functionalized with protein-bound ligands) for controlled protein release. Contrary to traditional hydrogels, which release proteins mainly through diffusion, affinity hydrogels stably retain and sustainably release proteins based mainly on diffusion coupled with a binding reaction. These hydrogels can also be modulated to release proteins in response to defined molecules in a triggered manner. Future research efforts may focus on the development of intelligent affinity hydrogels to mimic the properties of human tissues in sensing different environmental stimuli for on-demand release of single or multiple proteins (i.e., biomimetic intelligence for protein delivery).
Collapse
Affiliation(s)
- Lidya Abune
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
77
|
Sarrigiannidis S, Rey J, Dobre O, González-García C, Dalby M, Salmeron-Sanchez M. A tough act to follow: collagen hydrogel modifications to improve mechanical and growth factor loading capabilities. Mater Today Bio 2021; 10:100098. [PMID: 33763641 PMCID: PMC7973388 DOI: 10.1016/j.mtbio.2021.100098] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Collagen hydrogels are among the most well-studied platforms for drug delivery and in situ tissue engineering, thanks to their low cost, low immunogenicity, versatility, biocompatibility, and similarity to the natural extracellular matrix (ECM). Despite collagen being largely responsible for the tensile properties of native connective tissues, collagen hydrogels have relatively low mechanical properties in the absence of covalent cross-linking. This is particularly problematic when attempting to regenerate stiffer and stronger native tissues such as bone. Furthermore, in contrast to hydrogels based on ECM proteins such as fibronectin, collagen hydrogels do not have any growth factor (GF)-specific binding sites and often cannot sequester physiological (small) amounts of the protein. GF binding and in situ presentation are properties that can aid significantly in the tissue regeneration process by dictating cell fate without causing adverse effects such as malignant tumorigenic tissue growth. To alleviate these issues, researchers have developed several strategies to increase the mechanical properties of collagen hydrogels using physical or chemical modifications. This can expand the applicability of collagen hydrogels to tissues subject to a continuous load. GF delivery has also been explored, mathematically and experimentally, through the development of direct loading, chemical cross-linking, electrostatic interaction, and other carrier systems. This comprehensive article explores the ways in which these parameters, mechanical properties and GF delivery, have been optimized in collagen hydrogel systems and examines their in vitro or in vivo biological effect. This article can, therefore, be a useful tool to streamline future studies in the field, by pointing researchers into the appropriate direction according to their collagen hydrogel design requirements.
Collapse
Affiliation(s)
| | | | - O. Dobre
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8LT, UK
| | - C. González-García
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8LT, UK
| | - M.J. Dalby
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8LT, UK
| | - M. Salmeron-Sanchez
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8LT, UK
| |
Collapse
|
78
|
Khan AUR, Morsi Y, Zhu T, Ahmad A, Xie X, Yu F, Mo X. Electrospinning: An emerging technology to construct polymer-based nanofibrous scaffolds for diabetic wound healing. FRONTIERS OF MATERIALS SCIENCE 2021; 15:10-35. [DOI: 10.1007/s11706-021-0540-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 01/05/2025]
|
79
|
Khan AUR, Huang K, Khalaji MS, Yu F, Xie X, Zhu T, Morsi Y, Jinzhong Z, Mo X. Multifunctional bioactive core-shell electrospun membrane capable to terminate inflammatory cycle and promote angiogenesis in diabetic wound. Bioact Mater 2021; 6:2783-2800. [PMID: 33665509 PMCID: PMC7900605 DOI: 10.1016/j.bioactmat.2021.01.040] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/25/2021] [Accepted: 01/31/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetic wound (DW) healing is a major clinical challenge due to multifactorial complications leading to prolonged inflammation. Electrospun nanofibrous (NF) membranes, due to special structural features, are promising biomaterials capable to promote DW healing through the delivery of active agents in a controlled manner. Herein, we report a multifunctional composite NF membrane loaded with ZnO nanoparticles (NP) and oregano essential oil (OEO), employing a new loading strategy, capable to sustainedly co-deliver bioactive agents. Physicochemical characterization revealed the successful fabrication of loaded nanofibers with strong in vitro anti-bacterial and anti-oxidant activities. Furthermore, in vivo wound healing confirmed the potential of bioactive NF membranes in epithelialization and granulation tissue formation. The angiogenesis was greatly prompted by the bioactive NF membranes through expression of vascular endothelial growth factor (VEGF). Moreover, the proposed NF membrane successfully terminated the inflammatory cycle by downregulating the pro-inflammatory cytokines interleukin −6 (IL-6) and matrix metalloproteinases-9 (MMP-9). In vitro and in vivo studies revealed the proposed NF membrane is a promising dressing material for the healing of DW. A modified loading strategy was employed for dual bioactive agents through electrospinning. The nanofibers sustainedly released the two bioactive agents. The fabricated bioactive membranes turned out to be biocompatible, antioxidant and antibacterial. The proposed bioactive membranes have posesses the potential of healing diabetic wounds.
Collapse
Affiliation(s)
- Atta Ur Rehman Khan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Kai Huang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, PR China
| | - Mina Shahriari Khalaji
- Microbiological Engineering and Industrial Biotechnology Group, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Fan Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Xianrui Xie
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials of Shanghai University of Engineering Science, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, No. 333 Longteng Road, Shanghai, 201620, PR China
| | - Yosry Morsi
- Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, Boroondara, VIC, 3122, Australia
| | - Zhao Jinzhong
- Department of Sports Medicine, Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, PR China
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| |
Collapse
|
80
|
Rother S, Ruiz-Gómez G, Balamurugan K, Koehler L, Fiebig KM, Galiazzo VD, Hempel U, Moeller S, Schnabelrauch M, Waltenberger J, Pisabarro MT, Scharnweber D, Hintze V. Hyaluronan/Collagen Hydrogels with Sulfated Glycosaminoglycans Maintain VEGF165 Activity and Fine-Tune Endothelial Cell Response. ACS APPLIED BIO MATERIALS 2020; 4:494-506. [DOI: 10.1021/acsabm.0c01001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Gloria Ruiz-Gómez
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, Dresden 01307, Germany
| | | | - Linda Koehler
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Karen M. Fiebig
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Vanessa D. Galiazzo
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Ute Hempel
- Institute of Physiological Chemistry, Carl Gustav Carus Faculty of Medicine, TU Dresden, Fiedlerstraße 42, 01307 Dresden, Germany
| | - Stephanie Moeller
- Biomaterials Department, INNOVENT e.V., Prüssingstr. 27B, 07745 Jena, Germany
| | | | - Johannes Waltenberger
- Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - M. Teresa Pisabarro
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, Dresden 01307, Germany
| | - Dieter Scharnweber
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| |
Collapse
|
81
|
Li Z, Chen S, Wu B, Liu Z, Cheng L, Bao Y, Ma Y, Chen L, Tong X, Dai F. Multifunctional Dual Ionic-Covalent Membranes for Wound Healing. ACS Biomater Sci Eng 2020; 6:6949-6960. [DOI: 10.1021/acsbiomaterials.0c01512] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Zhi Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
| | - Sihao Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
| | - Baiqing Wu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Zulan Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
| | - Lan Cheng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Yu Bao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Yan Ma
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Lei Chen
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
| | - Xiaoling Tong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Textile and Garment, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| |
Collapse
|
82
|
Walimbe T, Panitch A. Best of Both Hydrogel Worlds: Harnessing Bioactivity and Tunability by Incorporating Glycosaminoglycans in Collagen Hydrogels. Bioengineering (Basel) 2020; 7:E156. [PMID: 33276506 PMCID: PMC7711789 DOI: 10.3390/bioengineering7040156] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 01/13/2023] Open
Abstract
Collagen, the most abundant protein in mammals, has garnered the interest of scientists for over 50 years. Its ubiquitous presence in all body tissues combined with its excellent biocompatibility has led scientists to study its potential as a biomaterial for a wide variety of biomedical applications with a high degree of success and widespread clinical approval. More recently, in order to increase their tunability and applicability, collagen hydrogels have frequently been co-polymerized with other natural and synthetic polymers. Of special significance is the use of bioactive glycosaminoglycans-the carbohydrate-rich polymers of the ECM responsible for regulating tissue homeostasis and cell signaling. This review covers the recent advances in the development of collagen-based hydrogels and collagen-glycosaminoglycan blend hydrogels for biomedical research. We discuss the formulations and shortcomings of using collagen in isolation, and the advantages of incorporating glycosaminoglycans (GAGs) in the hydrogels. We further elaborate on modifications used on these biopolymers for tunability and discuss tissue specific applications. The information presented herein will demonstrate the versatility and highly translational value of using collagen blended with GAGs as hydrogels for biomedical engineering applications.
Collapse
Affiliation(s)
- Tanaya Walimbe
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
- Department of Surgery, University of California Davis Health, Sacramento, CA 95817, USA
| |
Collapse
|
83
|
Guan N, Liu Z, Zhao Y, Li Q, Wang Y. Engineered biomaterial strategies for controlling growth factors in tissue engineering. Drug Deliv 2020; 27:1438-1451. [PMID: 33100031 PMCID: PMC7594870 DOI: 10.1080/10717544.2020.1831104] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/19/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Growth factors are multi-functional signaling molecules that coordinate multi-stage process of wound healing. During wound healing, growth factors are transmitted to wound environment in a positive and physiologically related way, therefore, there is a broad prospect for studying the mediated healing process through growth factors. However, growth factors (GFs) themselves have disadvantages of instability, short life, rapid inactivation of physiological conditions, low safety and easy degradation, which hinder the clinical use of GFs. Rapid development of delivery strategies for GFs has been trying to solve the instability and insecurity of GFs. Particularly, in recent years, GFs delivered by scaffolds based on biomaterials have become a hotspot in this filed. This review introduces various delivery strategies for growth factors based on new biodegradable materials, especially polysaccharides, which could provide guidance for the development of the delivery strategies for growth factors in clinic.
Collapse
Affiliation(s)
- Na Guan
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, P. R. China
| | - Zhihai Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, P. R. China
| | - Yonghui Zhao
- Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Qiu Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
84
|
Biofunctionalized fibrin gel co-embedded with BMSCs and VEGF for accelerating skin injury repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 121:111749. [PMID: 33579437 DOI: 10.1016/j.msec.2020.111749] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/07/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Rapid and effective repair of epithelial tissue is desirable for improving the success rate of operation and reducing postoperative complications. Hydrogel is a widely studied wound repair material, especially as a wound dressing for damaged epithelial tissue. Based on the catalytic effect of thrombin on fibrinogen, in this study, a three-dimensional fibrin gel which of adequate epithelial cell compatibility was constructed by using thrombin and fibrinogen under the cross-linking action of calcium ion. Immunofluorescence staining and hematoxylin-eosin (H&E) staining showed that bone marrow mesenchymal stem cell (BMSC) was embedded in fibrin gel. Furthermore, vascular endothelial growth factor (VEGF) was used to induce BMSC to differentiate into CD31+ and vWF+ endothelial cell (EC) in fibrin gel. The results showed that the fibrin gel surface may effectively promote the adhesion and proliferation of EC and smooth muscle cell (SMC). After 15 days of culture, it was found that the BMSC embedded in the hydrogel had differentiated into EC. The results of in vivo skin wound experiment in rats further proved that the fibrin gel containing BMSC could promote wound healing and repair, and showed the potential to promote neovascularization at the injured site. The construction method of hydrogel materials proposed in this study has potential application value in the field of regenerative medicine.
Collapse
|
85
|
Sharma A, Puri V, Kumar P, Singh I. Biopolymeric, Nanopatterned, Fibrous Carriers for Wound Healing Applications. Curr Pharm Des 2020; 26:4894-4908. [DOI: 10.2174/1381612826666200701152217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/11/2020] [Indexed: 12/15/2022]
Abstract
Background:
Any sort of wound injury leads to skin integrity and further leads to wound formation.
Millions of deaths are reported every year, which contributes to an economical hamper world widely, this accounts
for 10% of death rate that insight into various diseases.
Current Methodology:
Rapid wound healing plays an important role in effective health care. Wound healing is a
multi-factorial physiological process, which helps in the growth of new tissue to render the body with the imperative
barrier from the external environment. The complexity of this phenomenon makes it prone to several abnormalities.
Wound healing, as a normal biological inherent process occurs in the body, which is reaped through four
highly defined programmed phases, such as hemostasis, inflammation, proliferation, and remodeling and these
phases occur in the proper progression. An overview, types, and classification of wounds along with the stages of
wound healing and various factors affecting wound healing have been discussed systematically. Various biopolymers
are reported for developing nanofibers and microfibers in wound healing, which can be used as a therapeutic
drug delivery for wound healing applications. Biopolymers are relevant for biomedical purposes owing to
biodegradability, biocompatibility, and non- toxicity. Biopolymers such as polysaccharides, proteins and various
gums are used for wound healing applications. Patents and future perspectives have been given in the concluding
part of the manuscript. Overall, applications of biopolymers in the development of fibers and their applications in
wound healing are gaining interest in researchers to develop modified biopolymers and tunable delivery systems
for effective management and care of different types of wounds.
Collapse
Affiliation(s)
- Ameya Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vivek Puri
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
86
|
Cheng Y, Li Y, Huang S, Yu F, Bei Y, Zhang Y, Tang J, Huang Y, Xiang Q. Hybrid Freeze-Dried Dressings Composed of Epidermal Growth Factor and Recombinant Human-Like Collagen Enhance Cutaneous Wound Healing in Rats. Front Bioeng Biotechnol 2020; 8:742. [PMID: 32760705 PMCID: PMC7375021 DOI: 10.3389/fbioe.2020.00742] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/10/2020] [Indexed: 01/13/2023] Open
Abstract
Epidermal growth factor (EGF) is important for promoting skin repair and remodeling. Native collagen is also widely used as a scaffold for skin tissue engineering. The limitations of EGF include easy decomposition or inactivation, whereas native collagen is immunogenic and has poor solubility. Therefore, we constructed a freeze-dried dressing based on the recombinant human-like collagen (RHC) to act as a carrier for EGF (RHC/EGF freeze-dried dressing) and promote skin wound closure. Here, the freeze-dried dressing that combined EGF and RHC significantly enhanced the proliferation, adhesion, and spreading of NIH/3T3 fibroblasts and migration of HaCaT keratinocytes at the wound site. The physicochemical characteristics of the RHC/EGF freeze-dried dressing investigated using scanning electron microscopy, Fourier transform infrared (FTIR) spectroscopy, and differential scanning calorimetry revealed that it was a loose and porous cake that redissolved quickly. The molecular mechanisms involved in cell proliferation and angiogenesis were also assessed. The expression levels of the markers Ki-67, proliferating cell nuclear antigen, vascular endothelial growth factor, and cluster of differentiation 31 were significantly increased after treatment with the RHC/EGF freeze-dried dressing (P < 0.01, vs. RHC or EGF alone). This increase indicated that the RHC/EGF freeze-dried dressing significantly accelerated wound closure, re-epithelialization, and the orderly arrangement and deposition of collagen in the Sprague–Dawley rats with full-thickness skin defects. This work describes a significant step toward the development of wound environments conducive to healing, and the RHC/EGF freeze-dried dressing is a potential therapeutic strategy in wound management.
Collapse
Affiliation(s)
- Yating Cheng
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Yangfan Li
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Shiyi Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Fenglin Yu
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Yu Bei
- Biopharmaceutical R&D Center of Jinan University, Guangzhou, China
| | - Yifan Zhang
- Biopharmaceutical R&D Center of Jinan University, Guangzhou, China
| | - Jianzhong Tang
- Biopharmaceutical R&D Center of Jinan University, Guangzhou, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,Biopharmaceutical R&D Center of Jinan University, Guangzhou, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,Biopharmaceutical R&D Center of Jinan University, Guangzhou, China
| |
Collapse
|
87
|
Nurkesh A, Jaguparov A, Jimi S, Saparov A. Recent Advances in the Controlled Release of Growth Factors and Cytokines for Improving Cutaneous Wound Healing. Front Cell Dev Biol 2020; 8:638. [PMID: 32760728 PMCID: PMC7371992 DOI: 10.3389/fcell.2020.00638] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
Bioengineered materials are widely utilized due to their biocompatibility and degradability, as well as their moisturizing and antibacterial properties. One field of their application in medicine is to treat wounds by promoting tissue regeneration and improving wound healing. In addition to creating a physical and chemical barrier against primary infection, the mechanical stability of the porous structure of biomaterials provides an extracellular matrix (ECM)-like niche for cells. Growth factors (GFs) and cytokines, which are secreted by the cells, are essential parts of the complex process of tissue regeneration and wound healing. There are several clinically approved GFs for topical administration and direct injections. However, the limited time of bioactivity at the wound site often requires repeated drug administration that increases cost and may cause adverse side effects. The tissue regeneration promoting factors incorporated into the materials have significantly enhanced wound healing in comparison to bolus drug treatment. Biomaterials protect the cargos from protease degradation and provide sustainable drug delivery for an extended period of time. This prolonged drug bioactivity lowered the dosage, eliminated the need for repeated administration, and decreased the potential of undesirable side effects. In the following mini-review, recent advances in the field of single and combinatorial delivery of GFs and cytokines for treating cutaneous wound healing will be discussed.
Collapse
Affiliation(s)
- Ayan Nurkesh
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Alexandr Jaguparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Shiro Jimi
- Central Laboratory for Pathology and Morphology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
88
|
Feng X, Zhang X, Li S, Zheng Y, Shi X, Li F, Guo S, Yang J. Preparation of aminated fish scale collagen and oxidized sodium alginate hybrid hydrogel for enhanced full-thickness wound healing. Int J Biol Macromol 2020; 164:626-637. [PMID: 32668308 DOI: 10.1016/j.ijbiomac.2020.07.058] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/23/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022]
Abstract
Acute full-thickness wounds require a more extended healing period, thus increasing the risk of infection. Severe infection frequently resulted in wound ulceration, necrosis, and even life-threatening complications. Here, a hybrid hydrogel comprising aminated collagen (AC), oxidized sodium alginate (OSA), and antimicrobial peptides (polymyxin B sulfate and bacitracin) was developed to enhance full-thickness wound healing. The AC with low immunogenicity and high biocompatibility was made from marine fish scales, which are eco-friendly, low-cost, and sustainable. The cross-linked hydrogel was formed by a Schiff base reaction without any catalysts and additional procedures. As expected, the presented hybrid hydrogel can effectively against E. coli and S. aureus, as well as promote cell growth and angiogenesis in vitro. In addition, the hydrogel can promote full-thickness wound healing in a rat model through accelerating reepithelialization, collagen deposition, and angiogenesis. Our work demonstrated that the hybrid hydrogel has promising applications in the field of wound healing, which would prompt the utilization of marine fish resources during food processing.
Collapse
Affiliation(s)
- Xiaolian Feng
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Xiaofang Zhang
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Shiqi Li
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yunquan Zheng
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China.
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Feng Li
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Shaobin Guo
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China.
| |
Collapse
|
89
|
Amorim S, Reis CA, Reis RL, Pires RA. Extracellular Matrix Mimics Using Hyaluronan-Based Biomaterials. Trends Biotechnol 2020; 39:90-104. [PMID: 32654775 DOI: 10.1016/j.tibtech.2020.06.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
Abstract
Hyaluronan (HA) is a critical element of the extracellular matrix (ECM). The regulated synthesis and degradation of HA modulates the ECM chemical and physical properties that, in turn, influence cellular behavior. HA triggers signaling pathways associated with the adhesion, proliferation, migration, and differentiation of cells, mediated by its interaction with specific cellular receptors or by tuning the mechanical properties of the ECM. This review summarizes the recent advances on strategies used to mimic the HA present in the ECM to study healthy or pathological cellular behavior. This includes the development of HA-based 2D and 3D in vitro tissue models for the seeding and encapsulation of cells, respectively, and HA particles as carriers for the targeted delivery of therapeutic agents.
Collapse
Affiliation(s)
- Sara Amorim
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto, IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal; Department of Pathology and Oncology, Faculty of Medicine, Porto University, Porto, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo A Pires
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
90
|
Wathoni N, Rusdiana T, Hasanah AN, Pratama AR, Okajima M, Kaneko T, Mohammed AFA, Putera BW, Arima H. Epidermal growth factor in sacran hydrogel film accelerates fibroblast migration. J Adv Pharm Technol Res 2020; 11:74-80. [PMID: 32587820 PMCID: PMC7305779 DOI: 10.4103/japtr.japtr_147_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/16/2019] [Accepted: 02/13/2020] [Indexed: 01/09/2023] Open
Abstract
Epidermal growth factor (EGF) accelerates epidermal regeneration, and it is widely studied as a wound-healing agent. However, the special carrier for the topical administration of EGF is urgently needed to deliver EGF on the wound site. In a preceding study, sacran hydrogel film (Sac-HF) showed a possible use as a dressing material for wound healing, as well as a good capability as a drug carrier. In the current study, we prepared Sac-HF containing EGF (Sac/EGF-HF) and then characterized their physicochemical properties, including thickness, swelling ratio, degradability, tensile strength, and morphology. In addition, we have also conducted thermal and crystallography studies using differential scanning calorimetry (DSC) and X-ray diffraction, respectively. Furthermore, we investigated the in vitro influence of Sac/EGF-HF on cell migration using a fibroblast cell line. Morphology study confirmed that the casting method used for the film preparation resulted in a homogeneous film of Sac/EGF-HF. Furthermore, EGF significantly increased the thickness, tensile strength, and degradability of Sac/EGF-HF compared to Sac-HF. Sac/EGF-HF had a lower swelling ability compared to Sac-HF; this result corroborated the tensile strength result. Interestingly, X-ray diffraction and DSC results showed that Sac/EGF-HF had an amorphous shape. The in vitro studies revealed that Sac/EGF-HF induced the fibroblast migration activity. These results conclude that Sac/EGF-HF has the potential properties of HF for biomedical applications.
Collapse
Affiliation(s)
- Nasrul Wathoni
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Taofik Rusdiana
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Aliya Nur Hasanah
- Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Arvenda Rezky Pratama
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Maiko Okajima
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, Ishikawa, Japan
| | - Tatsuo Kaneko
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, Ishikawa, Japan
| | | | - Bayu Winata Putera
- Department of Operational, PT Prodia StemCell Indonesia, Jakarta, Indonesia
| | - Hidetoshi Arima
- Laboratory of Evidence-Based Pharmacotherapy, Daiichi University of Pharmacy, Fukuoka, Japan
| |
Collapse
|
91
|
Li J, Wu C, Chu PK, Gelinsky M. 3D printing of hydrogels: Rational design strategies and emerging biomedical applications. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2020; 140:100543. [DOI: 10.1016/j.mser.2020.100543] [Citation(s) in RCA: 350] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
92
|
Kalai Selvan N, Shanmugarajan T, Uppuluri VNVA. Hydrogel based scaffolding polymeric biomaterials: Approaches towards skin tissue regeneration. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101456] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
93
|
Li G, Han Q, Lu P, Zhang L, Zhang Y, Chen S, Zhang P, Zhang L, Cui W, Wang H, Zhang H. Construction of Dual-Biofunctionalized Chitosan/Collagen Scaffolds for Simultaneous Neovascularization and Nerve Regeneration. RESEARCH (WASHINGTON, D.C.) 2020; 2020:2603048. [PMID: 32851386 PMCID: PMC7436332 DOI: 10.34133/2020/2603048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/10/2020] [Indexed: 01/20/2023]
Abstract
Biofunctionalization of artificial nerve implants by incorporation of specific bioactive factors has greatly enhanced the success of grafting procedures for peripheral nerve regeneration. However, most studies on novel biofunctionalized implants have emphasized the promotion of neuronal and axonal repair over vascularization, a process critical for long-term functional restoration. We constructed a dual-biofunctionalized chitosan/collagen composite scaffold with Ile-Lys-Val-Ala-Val (IKVAV) and vascular endothelial growth factor (VEGF) by combining solution blending, in situ lyophilization, and surface biomodification. Immobilization of VEGF and IKVAV on the scaffolds was confirmed both qualitatively by staining and quantitatively by ELISA. Various single- and dual-biofunctionalized scaffolds were compared for the promotion of endothelial cell (EC) and Schwann cell (SC) proliferation as well as the induction of angiogenic and neuroregeneration-associated genes by these cells in culture. The efficacy of these scaffolds for vascularization was evaluated by implantation in chicken embryos, while functional repair capacity in vivo was assessed in rats subjected to a 10 mm sciatic nerve injury. Dual-biofunctionalized scaffolds supported robust EC and SC proliferation and upregulated the expression levels of multiple genes and proteins related to neuroregeneration and vascularization. Dual-biofunctionalized scaffolds demonstrated superior vascularization induction in embryos and greater promotion of vascularization, myelination, and functional recovery in rats. These findings support the clinical potential of VEGF/IKVAV dual-biofunctionalized chitosan/collagen composite scaffolds for facilitating peripheral nerve regeneration, making it an attractive candidate for repairing critical nerve defect. The study may provide a critical experimental and theoretical basis for the development and design of new artificial nerve implants with excellent biological performance.
Collapse
Affiliation(s)
- Guicai Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Qi Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Panjian Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Liling Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Yuezhou Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Shiyu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Ping Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Wenguo Cui
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, China
- Co-Innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Hongbo Zhang
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, 20520 Turku, Finland
| |
Collapse
|
94
|
Vijayan A, A S, Kumar GSV. PEG grafted chitosan scaffold for dual growth factor delivery for enhanced wound healing. Sci Rep 2019; 9:19165. [PMID: 31844069 PMCID: PMC6915706 DOI: 10.1038/s41598-019-55214-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Application of growth factors at wound site has improved the efficiency and quality of healing. Basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) induce proliferation of various cells in wound healing. Delivery of growth factor from controlled release systems protect it from degradation and also result in sustained delivery of it at the site of injury. The goal of the study was to develop a Polyethylene glycol (PEG) cross-linked cotton-like chitosan scaffold (CS-PEG-H) by freeze-drying method and chemically conjugate heparin to the scaffold to which the growth factors can be electrostatically bound and evaluate its wound healing properties in vitro and in vivo. The growth factor containing scaffolds induced increased proliferation of HaCaT cells, increased neovascularization and collagen formation seen by H and E and Masson's trichrome staining. Immunohistochemistry was performed using the Ki67 marker which increased proliferation of cells in growth factor containing scaffold treated group. Frequent dressing changes are a major deterrent to proper wound healing. Our system was found to release both VEGF and bFGF in a continuous manner and attained stability after 7 days. Thus our system can maintain therapeutic levels of growth factor at the wound bed thereby avoiding the need for daily applications and frequent dressing changes. Thus, it can be a promising candidate for wound healing.
Collapse
Affiliation(s)
- Amritha Vijayan
- Cancer Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
- Research Scholar, Department of Biotechnology, Faculty of Applied Science & Technology, University of Kerala, Trivandrum, Kerala, 695581, India
| | - Sabareeswaran A
- Histopathology laboratory, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, 695011, India
| | - G S Vinod Kumar
- Cancer Biology, Nano Drug Delivery Systems (NDDS), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
95
|
Dual Action of Sulfated Hyaluronan on Angiogenic Processes in Relation to Vascular Endothelial Growth Factor-A. Sci Rep 2019; 9:18143. [PMID: 31792253 PMCID: PMC6889296 DOI: 10.1038/s41598-019-54211-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/05/2019] [Indexed: 01/13/2023] Open
Abstract
Pathological healing characterized by abnormal angiogenesis presents a serious burden to patients’ quality of life requiring innovative treatment strategies. Glycosaminoglycans (GAG) are important regulators of angiogenic processes. This experimental and computational study revealed how sulfated GAG derivatives (sGAG) influence the interplay of vascular endothelial growth factor (VEGF)165 and its heparin-binding domain (HBD) with the signaling receptor VEGFR-2 up to atomic detail. There was profound evidence for a HBD-GAG-HBD stacking configuration. Here, the sGAG act as a “molecular glue” leading to recognition modes in which sGAG interact with two VEGF165-HBDs. A 3D angiogenesis model demonstrated the dual regulatory role of high-sulfated derivatives on the biological activity of endothelial cells. While GAG alone promote sprouting, they downregulate VEGF165-mediated signaling and, thereby, elicit VEGF165-independent and -dependent effects. These findings provide novel insights into the modulatory potential of sGAG derivatives on angiogenic processes and point towards their prospective application in treating abnormal angiogenesis.
Collapse
|
96
|
Kong X, Fu J, Shao K, Wang L, Lan X, Shi J. Biomimetic hydrogel for rapid and scar-free healing of skin wounds inspired by the healing process of oral mucosa. Acta Biomater 2019; 100:255-269. [PMID: 31606531 DOI: 10.1016/j.actbio.2019.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/18/2022]
Abstract
Inspired by the wound healing characteristics of the oral mucosa, a biomimetic hydrogel was prepared to realize the rapid and scar-free healing of skin wounds. Through monitoring the healing process of injured oral mucosa, we find out that the combination of high, rapid and sequential expression of some growth factors and the sterile-moist microenvironment are crucial for re-epithelialization and precise control of the inflammation process. On the base of our findings, a hydrogel loaded with several functional compounds was prepared to achieve a comprehensive simulation of the oral mucosal trauma microenvironment for skin wound healing. After 7 days treatment, the skin wound area of the treated group was only about 20% of that of the untreated group, and the proportion of collagen type III and type I in the treated group was much higher than that of the untreated group, suggesting lighter scar hyperplasia. The comprehensive treatment strategy of sequential expression of growth factors in combination with maintaining of a sterile and humid environment is expected to have great application prospect in the field of chronic trauma repair and cosmetic surgery. STATEMENT OF SIGNIFICANCE: Long healing time and scar hyperplasia during wound healing have been a serious problem in the past decades of wound healing research. Oral cavity wound healing occurs in an environment that sustains ongoing physical trauma and is rich in bacteria. Despite this, injuries to the mucosal surface often heal faster than cutaneous wounds and leave less noticeable scars. Therefore, in recent years, many scholars have begun to study the healing mechanism of oral mucosa, which supports a new inspiration for the study of skin wound repair: whether the injured skin can achieve a rapid scar-free healing effect similar to oral mucosa? Imitating the biological process of oral mucosa wound healing would be a promising therapeutic strategy in wound healing. Therefore, inspired by the wound healing characteristics of the oral mucosa, a biomimetic gel was prepared to realize the rapid and scar-free healing of skin wounds. Through monitoring the healing process of injured oral mucosa, the combination of high, rapid and sequential expression of some growth factors and sterile-moist microenvironment was crucial for re-epithelialization and precise control of the inflammation process. The comprehensive treatment strategy of sequential expression of growth factors in combination with maintance of a sterile and humid environment implies its potential use in the field of chronic trauma repair and cosmetic surgery.
Collapse
Affiliation(s)
- Xiaoying Kong
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China
| | - Jun Fu
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 Zhongguan West Road, Ningbo 315200, China
| | - Kai Shao
- Medical Experimental Center, Qilu Hospital of Shandong University (Qingdao), 758 Hefei Road, Qingdao 266035, China
| | - Lili Wang
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China
| | - Xuefang Lan
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China
| | - Jinsheng Shi
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China.
| |
Collapse
|
97
|
Abednejad A, Ghaee A, Morais ES, Sharma M, Neves BM, Freire MG, Nourmohammadi J, Mehrizi AA. Polyvinylidene fluoride-Hyaluronic acid wound dressing comprised of ionic liquids for controlled drug delivery and dual therapeutic behavior. Acta Biomater 2019; 100:142-157. [PMID: 31586728 DOI: 10.1016/j.actbio.2019.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 01/02/2023]
Abstract
To improve the efficacy of transdermal drug delivery systems, the physical and chemical properties of drugs need to be optimized to better penetrate into the stratum corneum and to better diffuse into the epidermis and dermis layers. Accordingly, dual-biological function ionic liquids composed of active pharmaceutical ingredients were synthesized, comprising both analgesic and anti-inflammatory properties, by combining a cation derived from lidocaine and anions derived from hydrophobic nonsteroidal anti-inflammatory drugs. Active pharmaceutical ingredient ionic liquids (API-ILs) were characterized through nuclear magnetic resonance, cytotoxicity assay, and water solubility assay. All properties were compared with those of the original drugs. By converting the analgesic and anti-inflammatory drugs into dual-function API-ILs, their water solubility increased up to 470-fold, without affecting their cytotoxic profile. These API-ILs were incorporated into a bilayer wound dressing composed of a hydrophobic polyvinylidene fluoride (PVDF) membrane to act as a drug reservoir and a biocompatible hyaluronic acid (HA) layer. The prepared bilayer wound dressing was characterized in terms of mechanical properties, membrane drug uptake and drug release behavior, and application in transdermal delivery, demonstrating to have desirable mechanical properties and improved release of API-ILs. The assessment of anti-inflammatory activity through the inhibition of LPS-induced production of nitric oxide and prostaglandin E2 by macrophages revealed that the prepared membranes containing API-ILs are as effective as those with the original drugs. Cell adhesion of fibroblasts on membrane surfaces and cell viability assay confirmed improved the viability and adhesion of fibroblasts on PVDF/HA membranes. Finally, wound healing assay performed with fibroblasts showed that the bilayer membranes containing dual-function API-ILs are not detrimental to wound healing, while displaying increased and controlled drug delivery and dual therapeutic behavior. STATEMENT OF SIGNIFICANCE: This work shows the preparation and characterization of bilayer wound dressings comprising dual-biological function active pharmaceutical ingredients based on ionic liquids with improved and controlled drug release and dual therapeutic efficiency. By converting analgesic and anti-inflammatory drugs into ionic liquids, their water solubility increases up to 470-fold. The prepared bilayer wound dressing membranes have desirable mechanical properties and improved release of drugs. The prepared membranes comprising ionic liquids display anti-inflammatory activity as effective as those with the original drugs. Cell adhesion of fibroblasts on membrane surfaces and cell viability assays show improved viability and adhesion of fibroblasts on PVDF/HA membranes, being thus of high relevance as effective transdermal drug delivery systems.
Collapse
|
98
|
Extracellular matrix-cell interactions: Focus on therapeutic applications. Cell Signal 2019; 66:109487. [PMID: 31778739 DOI: 10.1016/j.cellsig.2019.109487] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Extracellular matrix (ECM) macromolecules together with a multitude of different molecules residing in the extracellular space play a vital role in the regulation of cellular phenotype and behavior. This is achieved via constant reciprocal interactions between the molecules of the ECM and the cells. The ECM-cell interactions are mediated via cell surface receptors either directly or indirectly with co-operative molecules. The ECM is also under perpetual remodeling process influencing cell-signaling pathways on its part. The fragmentation of ECM macromolecules provides even further complexity for the intricate environment of the cells. However, as long as the interactions between the ECM and the cells are in balance, the health of the body is retained. Alternatively, any dysregulation in these interactions can lead to pathological processes and finally to various diseases. Thus, therapeutic applications that are based on retaining normal ECM-cell interactions are highly rationale. Moreover, in the light of the current knowledge, also concurrent multi-targeting of the complex ECM-cell interactions is required for potent pharmacotherapies to be developed in the future.
Collapse
|
99
|
Hachim D, Whittaker TE, Kim H, Stevens MM. Glycosaminoglycan-based biomaterials for growth factor and cytokine delivery: Making the right choices. J Control Release 2019; 313:131-147. [PMID: 31629041 PMCID: PMC6900262 DOI: 10.1016/j.jconrel.2019.10.018] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022]
Abstract
Controlled, localized drug delivery is a long-standing goal of medical research, realization of which could reduce the harmful side-effects of drugs and allow more effective treatment of wounds, cancers, organ damage and other diseases. This is particularly the case for protein "drugs" and other therapeutic biological cargoes, which can be challenging to deliver effectively by conventional systemic administration. However, developing biocompatible materials that can sequester large quantities of protein and release them in a sustained and controlled manner has proven challenging. Glycosaminoglycans (GAGs) represent a promising class of bio-derived materials that possess these key properties and can additionally potentially enhance the biological effects of the delivered protein. They are a diverse group of linear polysaccharides with varied functionalities and suitabilities for different cargoes. However, most investigations so far have focused on a relatively small subset of GAGs - particularly heparin, a readily available, promiscuously-binding GAG. There is emerging evidence that for many applications other GAGs are in fact more suitable for regulated and sustained delivery. In this review, we aim to illuminate the beneficial properties of various GAGs with reference to specific protein cargoes, and to provide guidelines for informed choice of GAGs for therapeutic applications.
Collapse
Affiliation(s)
- Daniel Hachim
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Thomas E Whittaker
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Hyemin Kim
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Molly M Stevens
- Department of Materials, Imperial College London, London, SW7 2AZ, United Kingdom; Department of Bioengineering, Imperial College London, London, SW7 2AZ, United Kingdom; Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom.
| |
Collapse
|
100
|
Chen YR, Zhou ZX, Zhang JY, Yuan FZ, Xu BB, Guan J, Han C, Jiang D, Yang YY, Yu JK. Low-Molecular-Weight Heparin-Functionalized Chitosan-Chondroitin Sulfate Hydrogels for Controlled Release of TGF-β3 and in vitro Neocartilage Formation. Front Chem 2019; 7:745. [PMID: 31737612 PMCID: PMC6839338 DOI: 10.3389/fchem.2019.00745] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
Repair of hyaline cartilage remains a huge challenge in clinic because of the avascular and aneural characteristics and the paucity of endogenous repair cells. Recently, tissue engineering technique, possessing unique capacity of repairing large tissue defects, avoiding donor complications and two-stage invasive surgical procedures, has been developed a promising therapeutic strategy for cartilage injury. In this study, we incorporated low-molecular-weight heparin (LMWH) into carboxymethyl chitosan-oxidized chondroitin sulfate (CMC-OCS) hydrogel for loading transforming growth factor-β3 (TGF-β3) as matrix of peripheral blood mesenchymal stem cells (PB-MSCs) to construct tissue-engineered cartilage. Meanwhile, three control hydrogels with or without LMWH and/or TGF-β3 were also prepared. The gelling time, microstructures, mechanical properties, degradation rate, cytotoxicity, and the release of TGF-β3 of different hydrogels were investigated. In vitro experiments evaluated the tri-lineage differentiation potential of PB-MSCs, combined with the proliferation, distribution, viability, morphology, and chondrogenic differentiation. Compared with non-LMWH-hydrogels, LMWH-hydrogels (LMWH-CMC-OCS-TGF-β3) have shorter gelling time, higher mechanical strength, slower degradation rate and more stable and lasting release of TGF-β3. After two weeks of culture in vitro, expression of cartilage-specific genes collagen type-2 (COL-2) and aggrecan (AGC), and secretion of glycosaminoglycan (GAG), and COL-2 proteins in LMWH-CMC-OCS-TGF-β3 group were significantly higher than those in other groups. COL-2 immunofluorescence staining showed that the proportion of COL-2 positive cells and immunofluorescence intensity in LMWH-CMC-OCS-TGF-β3 hydrogel were significantly higher than those in other groups. The LMWH-CMC-OCS-TGF-β3 hydrogel can slowly release TGF-β3 in a long term, and meanwhile the hydrogel can provide a biocompatible microenvironment for the growth and chondrogenic differentiation of PB-MSCs. Thus, LMWH functionalized CMC-OCS hydrogels proposed in this work will be beneficial for constructing functional scaffolds for tissue-engineered cartilage.
Collapse
Affiliation(s)
- You-Rong Chen
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Zhu-Xing Zhou
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Ji-Ying Zhang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Fu-Zhen Yuan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Bing-Bing Xu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Jian Guan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Chao Han
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China.,School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Dong Jiang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Yan-Yu Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,College of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
| | - Jia-Kuo Yu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|