Graphene Oxide-Embedded Extracellular MatrixDerived Hydrogel as a Multiresponsive Platform for 3D Bioprinting Applications

Authors

  • Laura Rueda-Gensini Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, 11171, Colombia
  • Julian A. Serna Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, 11171, Colombia
  • Javier Cifuentes Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, 11171, Colombia
  • Juan C. Cruz Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, 11171, Colombia
  • Carolina Muñoz-Camargo Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, 11171, Colombia

DOI:

https://doi.org/10.18063/ijb.v7i3.353

Keywords:

Extracellular matrix bioink, Graphene oxide, Reduced graphene oxide, Electroconductive hydrogel, Photocrosslinking

Abstract

Decellularized extracellular matrices (dECMs) have shown enormous potential for the biofabrication of tissues due to their biomimetic properties that promote enhanced cellular interaction and tissue regeneration. However, biofabrication schemes requiring electrostimulation pose an additional constraint due to the insulating properties of natural materials. Here, we propose a methacryloyl-modified decellularized small intestine submucosa (SISMA) hydrogel, embedded with graphene oxide (GO) nanosheets, for extrusion-based 3D bioprinting applications that require electrostimulation. Methacryloyl biochemical modification is performed to enhance the mechanical stability of dECM constructs by mediating photo-crosslinking reactions, and a multistep fabrication scheme is proposed to harness the bioactive and hydrophilic properties of GO and electroconductive properties of reduced GO. For this, GO was initially dispersed in SISMA hydrogels by exploiting its hydrophilicity and protein adsorption capabilities, and in situ reduction was subsequently performed to confer electroconductive abilities. SISMA-GO composite hydrogels were successfully prepared with enhanced structural characteristics, as shown by the higher crosslinking degree and increased elastic response upon blue-light exposure. Moreover, GO was homogeneously dispersed without affecting photocrosslinking reactions and hydrogel shear-thinning properties. Human adipose-derived mesenchymal stem cells were successfully bioprinted in SISMA-GO with high cell viability after 1 week and in situ reduction of GO during this period enhanced the electrical conductivity of these nanostructures. This work demonstrates the potential of SISMA-GO bioinks as bioactive and electroconductive scaffolds for electrostimulation applications in tissue engineering and regenerative medicine.

References

Kim B, Das S, Jang J, et al., 2020, Decellularized Extracellular Matrix-based Bioinks for Engineering Tissue and Organs pecific Microenvironments. Chem Rev, 120:10608–61.

Ng W, Chua C, Shen Y, 2019, Print Me An Organ! Why We Are Not There Yet, Prog Polym Sci, 97:101145.

Frantz C, Stewart KM, Weaver VM, 2010, The Extracellular Matrix at a Glance, J Cell Sci, 123:4195–200.

Schaefer L, Schaefer RM, 2009, Proteoglycans: From Structural Compounds to Signaling Molecules, Cell Tissue Res, 339:237–46.

Kim BS, Kim H, Gao G, et al., 2017, Decellularized Extracellular Matrix: A Step Towards the Next Generation Source for Bioink Manufacturing. Biofabrication, 9:034104.

Kohane DS, Langer R, 2008, Polymeric Biomaterials in Tissue Engineering. Pediatr Res, 63:487–91.

Badylak SF, 2002, The Extracellular Matrix as a Scaffold for Tissue Reconstruction. Semin Cell Dev Biol, 13:377–83.

Saldin LT, Cramer MC, Velankar SS, et al, 2017, Extracellular Matrix Hydrogels from Decellularized Tissues: Structure and Function. Acta Biomater, 49:1–15.

Pouliot RA, Young BM, Link PA, et al. Porcine Lung-Derived Extracellular Matrix Hydrogel Properties Are Dependent on Pepsin Digestion Time. Tissue Eng C Methods, 26:332–46.

Yang C, 2012, Enhanced physicochemical properties of collagen by using EDC/NHS-crosslinking. Bull Mater Sci, 35:913–8.

Curley CJ, Dolan EB, Otten M, et al., 2018, An Injectable Alginate/Extra Cellular Matrix (ECM) Hydrogel Towards Acellular Treatment of Heart Failure. Drug Deliv Transl Res, 9:1–13.

Choi B, Kim S, Lin B, et al., 2014, Cartilaginous Extracellular Matrix-Modified Chitosan Hydrogels for Cartilage Tissue Engineering. ACS Appl Mater Interfaces, 6:20110–21.

Ali M, Kumar A Pr, Yoo JJ, et al., 2019, A Photo-Crosslinkable Kidney ECM-Derived Bioink Accelerates Renal Tissue Formation. Adv Healthc Mater, 8:1800992.

Sionkowska A, Skopinska-Wisniewska J, Gawron M, et al., 2010, Chemical and Thermal Cross-linking of Collagen and Elastin Hydrolysates. Int J Biol Macromol, 47:570–7.

Davidenko N, Schuster CF, Bax DV, et al., 2015, Control of Crosslinking for Tailoring Collagen-based Scaffolds Stability and Mechanics. Acta Biomater, 25:131–42.

Reddy N, Reddy R, Jiang Q, 2015, Crosslinking Biopolymers for Biomedical Applications. Trends Biotechnol, 33:362–9.

O’Connell CD, Zhang B, Onofrillo C, et al., 2018, Tailoring the mechanical properties of gelatin methacryloyl hydrogels through manipulation of the photocrosslinking conditions. Soft Matter, 14:2142–51.

Ashwin S, Reed G, Michael M, 2016, Myelinated 3D Neural Culture Platform as a Physiological Model of Diabetic Peripheral Neuropathy. Front Bioeng Biotechnol, 4:02188.

Williams CG, Malik AN, Kim TK, et al., 2005, Variable Cytocompatibility of Six Cell Lines with Photoinitiators Used for Polymerizing Hydrogels and Cell Encapsulation. Biomaterials, 26:1211–8.

Diamantides N, Wang L, Pruiksma T, et al., 2017, Correlating Rheological Properties and Printability of Collagen Bioinks: The Effects of Riboflavin Photocrosslinking and pH. Biofabrication, 9:034102.

Parthiban SP, Athirasala A, Tahayeri A, et al., 2020, BoneMA Synthesis and Characterization of a Methacrylated Bonederived Hydrogel for Bioprinting of Vascularized Tissues. BioRxiv, 2020:974063.

Kim SH, Yeon YK, Lee JM, et al., 2018, Precisely Printable and Biocompatible Silk Fibroin Bioink for Digital Light Processing 3D Printing. Nat Commun, 9:1620.

Poldervaart MT, Goversen B, de Ruijter M, et al., 2017, 3D Bioprinting of Methacrylated Hyaluronic Acid (MeHA) Hydrogel with Intrinsic Osteogenicity. PLoS One, 12:e0177628.

Paxton N, Smolan W, Böck T, et al., 2017, Proposal to Assess Printability of Bioinks for Extrusion-based Bioprinting and Evaluation of Rheological Properties Governing Bioprintability. Biofabrication, 9:044107.

Li J, Chen M, Fan X, et al, 2016, Recent Advances in Bioprinting Techniques: Approaches, Applications and Future Prospects. J Transl Med, 14:271.

Derakhshanfar S, Mbeleck R, Xu K, et al., 2018, 3D Bioprinting for Biomedical Devices and Tissue Engineering: A Review of Recent Trends and Advances. Bioact Mater, 3:144–56.

Agarwala S, 2020, Electrically Conducting Hydrogels for Health care: Concept Fabrication Methods, and Applications. Int J Bioprint, 6:273.

Thakral G, LaFontaine J, Najafi B, et al., 2013, Electrical Stimulation to Accelerate Wound Healing. Diabetic Foot Ankle, 4:22081.

Ashrafi M, Alonso-Rasgado T, Baguneid M, et al., 2017, The Efficacy of Electrical Stimulation in Lower Extremity Cutaneous Wound Healing: A Systematic Review. Exp Dermatol, 26:171–8.

Meng S, Rouabhia M, Zhang Z, 2012, Electrical Stimulation Modulates Osteoblast Proliferation and Bone Protein Production through Heparin-Bioactivated Conductive Scaffolds. Bioelectromagnetics, 34:189–99.

Leppik L, Zhihua H, Mobini S, et al., 2018, Combining Electrical Stimulation and Tissue Engineering to Treat Large Bone Defects in a Rat Model. Sci Rep, 8:6307.

Chen C, Bai X, Ding Y, et al., 2019, Electrical Stimulation as a Novel Tool for Regulating Cell Behavior in Tissue Engineering. Biomater Res, 23:25.

Fu C, Pan S, Ma Y, et al., 2019, Effect of Electrical Stimulation Combined with Graphene-oxide-based Membranes on Neural Stem Cell Proliferation and Differentiation. Artif Cells Nanomed Biotechnol, 47:1867–76.

Kwon HJ, Lee GS, Chun H, 2016, Electrical Stimulation Drives Chondrogenesis of Mesenchymal Stem Cells in the Absence of Exogenous Growth Factors. Sci Rep, 6:39302.

Sadeghian RB, Ebrahimi M, Salehi S, 2017, Electrical Stimulation of Microengineered Skeletal Muscle Tissue: Effect of Stimulus Parameters on Myotube Contractility and Maturation. J Tissue Eng Regen Med, 12:912–22.

Hirt MN, Boeddinghaus J, Mitchell A, et al., 2014, Functional Improvement and Maturation of Rat and Human Engineered Heart Tissue by Chronic Electrical Stimulation. J Mol Cell Cardiol, 74:151–61.

Ashtari K, Nazari H, Ko H, et al., 2019, Electrically Conductive Nanomaterials for Cardiac Tissue Engineering. Adv Drug Deliv Rev, 144:162–79.

Javadi M, Gu Q, Naficy S, et al., 2017, Conductive Tough Hydrogel for Bioapplications. Macromol Biosci, 18:1700270.

Zhou X, Cui H, Zhang LG, et al., 2017, 3D Bioprinted Graphene Oxide-incorporated Matrix for Promoting Chondrogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells. Carbon, 116:615–24.

Ku SH, Park CB, 2013, Myoblast Differentiation on Graphene Oxide. Biomaterials, 34:2017–23.

Liu Y, Zhang B, Xu Q, et al., 2018, Development of Graphene Oxide/Polyaniline Inks for High Performance Flexible Microsupercapacitors via Extrusion Printing. Adv Funct Mater, 28:1706592.

Sánchez-Palencia DM, D’Amore A, González-Mancera A, et al., 2014, Effects of Fabrication on the Mechanics Microstructure and Micromechanical Environment of Small Intestinal Submucosa Scaffolds for Vascular Tissue Engineering. J Biomech, 47:2766–73.

Marcano DC, Kosynkin DV, Berlin JM, et al., 2010, Improved Synthesis of Graphene Oxide. ACS Nano, 4:4806–14.

Gaudet ID, Shreiber DI, 2012, Characterization of Methacrylated Type-I Collagen as a Dynamic Photoactive Hydrogel. Biointerphases, 7:25.

Shih CJ, Lin S, Sharma R, et al., 2011, Understanding the Ph Dependent Behavior of Graphene Oxide Aqueous Solutions: A Comparative Experimental and Molecular Dynamics Simulation Study. Langmuir, 28:235–41.

Capella-Monsonís H, Coentro JQ, Graceffa V, et al., 2018, An Experimental Toolbox for Characterization of Mammalian Collagen Type I in Biological Specimens. Nat Protoc, 13:507–29.

Linero I, Chaparro O, 2014, Paracrine Effect of Mesenchymal Stem Cells Derived from Human Adipose Tissue in Bone Regeneration. PLoS One, 9:e107001.

Hermanson GT, 2013, Zero-Length Crosslinkers. In: Bioconjugate Techniques, Elsevier, Amsterdam, Netherlands, p259–73.

Lim KS, Schon BS, Mekhileri NV, et al., 2016, New Visible- Light Photoinitiating System for Improved Print Fidelity in Gelatin-Based Bioinks. ACS Biomater Sci Eng, 2:1752–62.

Yoon HJ, Shin SR, Cha JM, et al., 2016, Cold Water Fish Gelatin Methacryloyl Hydrogel for Tissue Engineering Application. PLoS One, 11:e0163902.

Drzewiecki KE, Parmar AS, Gaudet ID, et al., 2014, Methacrylation Induces Rapid Temperature-Dependent, Reversible Self-Assembly of Type-I Collagen. Langmuir, 30:11204–11.

Ravichandran R, Islam MM, Alarcon EI, et al., 2016, Functionalised Type-I Collagen as a Hydrogel Building Block for Bio-orthogonal Tissue Engineering Applications. J Mater Chem B, 4:318–26.

Tang H, Zhang S, Huang T, et al., 2020, Effects of pH and Electrolytes on the Sheet-to-Sheet Aggregation Mode of Graphene Oxide in Aqueous Solutions. Environ Sci Nano, 7:984–95.

Hong BJ, Compton OC, An Z, et al., 2011, Successful Stabilization of Graphene Oxide in Electrolyte Solutions: Enhancement of Biofunctionalization and Cellular Uptake. ACS Nano, 6:63–73.

Cha C, Shin SR, Gao X, et al., 2013, Controlling Mechanical Properties of Cell-Laden Hydrogels by Covalent Incorporation of Graphene Oxide. Small, 10:514–23.

Huang CT, Shrestha LK, Ariga K, et al., 2017, A Graphene polyurethane Composite Hydrogel as a Potential Bioink for 3D Bioprinting and Differentiation of Neural Stem Cells. J Mater Chem B, 5:8854–64.

Li Z, Shen J, Ma H, et al., 2012, Preparation and Characterization of pH and Temperature-Responsive Hydrogels with Surface-functionalized Graphene Oxide as the Crosslinker. Soft Matter, 8:3139.

Sayyar S, Murray E, Thompson BC, et al., 2015, Processable conducting graphene/chitosan hydrogels for tissue engineering. J Mater Chem B, 3:481–90.

Liu S, Mou S, Zhou C, et al., 2018, Off-the-Shelf Biomimetic Graphene OxideCollagen Hybrid Scaffolds Wrapped with Osteoinductive Extracellular Matrix for the Repair of Cranial Defects in Rats. ACS Appl Mater Interfaces, 10: 42948–58.

Rahman MM, Byanju B, Grewell D, et al., 2020, High-power Sonication of Soy Proteins: Hydroxyl Radicals and their Effects on Protein Structure. Ultrason Sonochem, 64:105019.

Fratzl P, 2008, Collagen: Structure and mechanics an introduction. In: Collagen. Springer, United States, p1–13.

Mane NC, Ponrathnam S, 2016, Effect of Chemical Crosslinking on Properties of Polymer Microbeads: A Review. Can Chem Trans, 3:473–85.

Huang R, Choe E, Min DB, 2006, Kinetics for Singlet Oxygen Formation by Riboflavin Photosensitization and the Reaction between Riboflavin and Singlet Oxygen. J Food Sci, 69:C726–32.

Jiang T, Munguia-Lopez JG, Flores-Torres S, et al., 2019, Extrusion Bioprinting of Soft Materials: An Emerging Technique for Biological Model Fabrication. Appl Phys Rev, 6:011310.

Wilson SA, Cross LM, Peak CW, et al., 2017, Shear-Thinning and Thermo-Reversible Nanoengineered Inks for 3D Bioprinting. ACS Appl Mater Interfaces, 9:43449–58.

Liu F, Gao Y, Li H, et al., 2014, Interaction of Propidium Iodide with Graphene Oxide and its Application for Live Cell Staining. Carbon, 71: 190–5.

Mironi-Harpaz I, Wang DY, Venkatraman S, et al., 2012, Photopolymerization of Cell-encapsulating Hydrogels: Crosslinking Efficiency Versus Cytotoxicity. Acta Biomater, 8:1838–48.

Lock JG, Wehrle-Haller B, Strömblad S, 2008, Cell-Matrix Adhesion Complexes: Master Control Machinery of Cell Migration. Semin Cancer Biol, 18:65–76.

Kumar S, Parekh SH, 2020, Linking Graphene-based Material Physicochemical Properties with Molecular Adsorption Structure and Cell Fate. Commun Chem, 3:8.

Singh RK, Kumar R, Singh DP, 2016, Graphene Oxide: Strategies for Synthesis Reduction and Frontier Applications. RSC Adv, 6:64993–5011.

Choi KM, Seo YK, Yoon HH, et al., 2008, Effect of Ascorbic Acid on Bone Marrow-derived Mesenchymal Stem Cell Proliferation and Differentiation. J Biosci Bioeng, 105:586–94.

Khalili D, 2016, Graphene Oxide: A Promising Carbocatalyst for the Regioselective Thiocyanation of Aromatic Amines Phenols, Anisols and Enolizable Ketones by Hydrogen Peroxide/KSCN in Water. New J Chem, 40:2547–53.

Wu JB, Lin ML, Cong X, et al., 2018, Raman Spectroscopy of Graphene-Based Materials and its Applications in Related Devices. Chem Soc Rev, 47:1822–73.

Muzyka R, Drewniak S, Pustelny T, et al., 2018, Characterization of Graphite Oxide and Reduced Graphene Oxide Obtained from Different Graphite Precursors and Oxidized by Different Methods Using Raman Spectroscopy. Materials, 11:1050.

Marsden AJ, Papageorgiou DG, Vallés C, et al., 2018, Electrical Percolation in Graphene-polymer Composites. 2D Materials, 5:032003.

Rao S, Upadhyay J, Polychronopoulou K, et al., 2018, Reduced Graphene Oxide: Effect of Reduction on Electrical Conductivity. J Compos Sci, 2:25.

Mohan VB, Brown R, Jayaraman K, et al., 2015, Characterisation of Reduced Graphene Oxide: Effects of Reduction Variables on Electrical Conductivity. Mater Sci Eng B, 193:49–60.

Kim M, Lee C, Jang J, 2013, Fabrication of Highly Flexible Scalable, and High-Performance Supercapacitors Using Polyaniline/Reduced Graphene Oxide Film with Enhanced Electrical Conductivity and Crystallinity. Adv Funct Mater, 24:2489–99.

Lang Q, Wu Y, Ren Y, et al., 2015, AC Electrothermal Circulatory Pumping Chip for Cell Culture. ACS Appl Mater Interfaces, 7:26792–801.

Adams TN, Turner PA, Janorkar AV, et al., 2014, Characterizing the Dielectric Properties of Human Mesenchymal Stem Cells and the Effects of Charged Elastin- Like Polypeptide Copolymer Treatment. Biomicrofluidics, 8: 054109.

Cho S, Thielecke H, 2008, Electrical Characterization of Human Mesenchymal Stem Cell Growth on Microelectrode. Microelectron Eng, 85:1272–4.

Walker BW, Lara RP, Mogadam E, et al., 2019, Rational Design of Microfabricated Electroconductive Hydrogels for Biomedical Applications. Prog Polym Sci, 92:135–57.

Barlian A, Judawisastra H, Alfarafisa NM, et al, 2018, Chondrogenic Differentiation of Adipose-derived Mesenchymal Stem Cells Induced by L-Ascorbic Acid and Platelet Rich Plasma on Silk Fibroin Scaffold. PeerJ, 6:e5809.

Kumar A, Nune KC, Misra RD, 2016, Understanding the Response of Pulsed Electric Field on Osteoblast Functions in Three-dimensional Mesh Structures. J Biomater Appl, 31:594–605.

Ross CL, 2016, The Use of Electric Magnetic, and Electromagnetic Field for Directed Cell Migration and Adhesion in Regenerative Medicine. Biotechnol Prog, 33:5–16.

Downloads

Published

2021-05-11