1
|
Wawszczak A, Kocki J, Kołodyńska D. Alginate as a Sustainable and Biodegradable Material for Medical and Environmental Applications-The Case Studies. J Biomed Mater Res B Appl Biomater 2024; 112:1-23. [PMID: 39269132 DOI: 10.1002/jbm.b.35475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Alginates are salts of alginic acid derived mainly from sea algae of the genus brown algae. They are also synthesized by some bacteria. They belong to negatively charged polysaccharides exhibiting some rheological properties. High plasticity and the ability to modify the structure are the reasons for their application in numerous industries. Moreover, when in contact with the living tissue, they do not trigger an immune response, and for this reason they are the most often tested materials for medical applications. The paper discusses the latest applications, including 3D bioprinting, drug delivery systems, and sorptive properties. Recognizing alginates as biomaterials, it emphasizes the necessity for precise processing and modification to industrialize them for specific uses. This review aims to provide a thorough understanding of the advancements in alginate research, underscoring their potential for innovative applications.
Collapse
Affiliation(s)
- Alicja Wawszczak
- Department of Inorganic Chemistry, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Dorota Kołodyńska
- Department of Inorganic Chemistry, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
2
|
Mrówczyńska E, Machalica K, Mazur AJ. Non-integrin laminin receptor (LamR) plays a role in axonal outgrowth from chicken DRG via modulating the Akt and Erk signaling. Front Cell Dev Biol 2024; 12:1433947. [PMID: 39144252 PMCID: PMC11322362 DOI: 10.3389/fcell.2024.1433947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
37/67 kDa laminin receptor (LamR)/ribosomal protein SA exhibits dual function as both a ribosomal protein and cell surface receptor for laminin. LamR influences critical cellular processes such as invasion, adhesion, and migration when acting as a receptor. Despite the acknowledged importance of LamR/67LR in various cellular processes, its contribution to the peripheral nervous system development is obscure. Thus, this study investigated the biological activity of LamR in peripheral axonal outgrowth in the presence of laminin-1 or Ile-Lys-Val-Ala-Val (IKVAV) peptide, whose important role in dorsal root ganglia (DRG) axonal outgrowth we recently showed. Unexpectedly, we did not observe LamR on the surface of DRG cells or in a conditioned medium, suggesting its intracellular action in the negative regulation of DRG axonal outgrowth. Using C-terminus LamR-targeting IgG, we demonstrated the role of LamR in that process, which is independent of the presence of Schwann cell precursors (SCPs) and is mediated by extracellular signal-regulated kinase (Erk) and Protein kinase B (Akt1/2/3) signaling pathways. Additionally, we show that the action of LamR towards laminin-1-dependent axonal outgrowth is unmasked only when the activity of integrin β1 is perturbed. We believe that modulation of LamR activity provides the basis for its use for inhibiting axon growth as a potential therapeutic agent for regulating abnormal or excessive neurite growth during neurodevelopmental diseases or pathological nerve regeneration.
Collapse
Affiliation(s)
- Ewa Mrówczyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | | | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| |
Collapse
|
3
|
Suhar RA, Huang MS, Navarro RS, Aviles Rodriguez G, Heilshorn SC. A Library of Elastin-like Proteins with Tunable Matrix Ligands for In Vitro 3D Neural Cell Culture. Biomacromolecules 2023; 24:5926-5939. [PMID: 37988588 DOI: 10.1021/acs.biomac.3c00941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Hydrogels with encapsulated cells have widespread biomedical applications, both as tissue-mimetic 3D cultures in vitro and as tissue-engineered therapies in vivo. Within these hydrogels, the presentation of cell-instructive extracellular matrix (ECM)-derived ligands and matrix stiffness are critical factors known to influence numerous cell behaviors. While individual ECM biopolymers can be blended together to alter the presentation of cell-instructive ligands, this typically results in hydrogels with a range of mechanical properties. Synthetic systems that allow for the facile incorporation and modulation of multiple ligands without modification of matrix mechanics are highly desirable. In the present work, we leverage protein engineering to design a family of xeno-free hydrogels (i.e., devoid of animal-derived components) consisting of recombinant hyaluronan and recombinant elastin-like proteins (ELPs), cross-linked together with dynamic covalent bonds. The ELP components incorporate cell-instructive peptide ligands derived from ECM proteins, including fibronectin (RGD), laminin (IKVAV and YIGSR), collagen (DGEA), and tenascin-C (PLAEIDGIELTY and VFDNFVL). By carefully designing the protein primary sequence, we form 3D hydrogels with defined and tunable concentrations of cell-instructive ligands that have similar matrix mechanics. Utilizing this system, we demonstrate that neurite outgrowth from encapsulated embryonic dorsal root ganglion (DRG) cultures is significantly modified by cell-instructive ligand content. Thus, this library of protein-engineered hydrogels is a cell-compatible system to systematically study cell responses to matrix-derived ligands.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- The Institute for Chemistry, Stanford University, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford, California 94305, United States
| | - Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Giselle Aviles Rodriguez
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
4
|
De S, Singh N. Collagen-alginate 3D microscaffolds for studying cellular migration. Int J Biol Macromol 2023; 245:125308. [PMID: 37315661 DOI: 10.1016/j.ijbiomac.2023.125308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/08/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
Metastasis is one of the major causes for cancer mortality. Its early steps comprise of invasion of basement membrane and migration. Thus, it is hypothesized that a platform, that allows quantification and grading of migration capability of cells can potentially be used for predicting metastatic potential. Two-dimensional (2D) models have been rendered inadequate for modelling in-vivo microenvironment due to various reasons. To attenuate homogeneity observed in 2D, three-dimensional (3D) platforms supplemented with bioinspired components have been designed. Unfortunately, till date there are no simple models to capture the migration of cells in 3D along with quantification of the process. In this study, we report an alginate-collagen based 3D model system, which can predict the migratory property of the cells within 72 h. The micron size of the scaffold enabled faster readout and the optimum pore-size provided conducive cellular growth environment. The platform's ability to allow observation of cellular migration was validated by encapsulating cells with transiently upregulated matrix metalloprotease 9 (MMP9), which has been reported to play a significant role in migration of cells during metastasis. The readout for migration was clustering of cells in the microscaffolds detected in a short span of 48 h. The observed clustering in MMP9 upregulated cells was validated by observing changes in the epithelial-mesenchymal transition (EMT) markers. Thus, this simple 3D platform can be used to study migration and predict the metastatic potential of cells.
Collapse
Affiliation(s)
- Shreemoyee De
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India; Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
5
|
Adamiak K, Sionkowska A. State of Innovation in Alginate-Based Materials. Mar Drugs 2023; 21:353. [PMID: 37367678 PMCID: PMC10302983 DOI: 10.3390/md21060353] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
This review article presents past and current alginate-based materials in each application, showing the widest range of alginate's usage and development in the past and in recent years. The first segment emphasizes the unique characteristics of alginates and their origin. The second segment sets alginates according to their application based on their features and limitations. Alginate is a polysaccharide and generally occurs as water-soluble sodium alginate. It constitutes hydrophilic and anionic polysaccharides originally extracted from natural brown algae and bacteria. Due to its promising properties, such as gelling, moisture retention, and film-forming, it can be used in environmental protection, cosmetics, medicine, tissue engineering, and the food industry. The comparison of publications with alginate-based products in the field of environmental protection, medicine, food, and cosmetics in scientific articles showed that the greatest number was assigned to the environmental field (30,767) and medicine (24,279), whereas fewer publications were available in cosmetic (5692) and food industries (24,334). Data are provided from the Google Scholar database (including abstract, title, and keywords), accessed in May 2023. In this review, various materials based on alginate are described, showing detailed information on modified composites and their possible usage. Alginate's application in water remediation and its significant value are highlighted. In this study, existing knowledge is compared, and this paper concludes with its future prospects.
Collapse
Affiliation(s)
- Katarzyna Adamiak
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarin 7 Street, 87-100 Torun, Poland;
- WellU sp.z.o.o., Wielkopolska 280, 81-531 Gdynia, Poland
| | - Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarin 7 Street, 87-100 Torun, Poland;
- Faculty of Health Sciences, Calisia University, Nowy Świat 4, 62-800 Kalisz, Poland
| |
Collapse
|
6
|
Jarrah RM, Potes MDA, Vitija X, Durrani S, Ghaith AK, Mualem W, Zamanian C, Bhandarkar AR, Bydon M. Alginate hydrogels: A potential tissue engineering intervention for intervertebral disc degeneration. J Clin Neurosci 2023; 113:32-37. [PMID: 37159956 DOI: 10.1016/j.jocn.2023.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 05/11/2023]
Abstract
Intervertebral disc (IVD) degeneration is a major cause of low back pain and disability, affecting millions of people worldwide. Current treatments for IVD degeneration are limited to invasive surgery or pain management. Recently, there has been increasing interest in the use of biomaterials, such as alginate hydrogels, for the treatment of IVD degeneration. Alginate hydrogels are an example of such a biomaterial that is biocompatible and can be tailored to mimic the native extracellular matrix of the IVD. Derived from alginate, a naturally derived polysaccharide from brown seaweed that can be transformed into a gelatinous solution, alginate hydrogels are emerging in the field of tissue engineering. They can be used to deliver therapeutic agents, such as growth factors or cells, to the site of injury, providing a localized and sustained release that may enhance treatment outcomes. This paper provides an overview on the use of alginate hydrogels for the treatment of IVD degeneration. We discuss the properties of alginate hydrogels and their potential applications for IVD regeneration, including the mechanism against IVD degeneration. We also highlight the research outcomes to date along with the challenges and limitations of using alginate hydrogels for IVD regeneration, including their mechanical properties, biocompatibility, and surgical compatibility. Overall, this review paper aims to provide a comprehensive overview of the current research on alginate hydrogels for IVD degeneration and to identify future directions for research in this area.
Collapse
Affiliation(s)
- Ryan M Jarrah
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Maria D Astudillo Potes
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Xheneta Vitija
- Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA; College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Sulaman Durrani
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Abdul Karim Ghaith
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - William Mualem
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Cameron Zamanian
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Archis R Bhandarkar
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Mohamad Bydon
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
7
|
Hebisch M, Klostermeier S, Wolf K, Boccaccini AR, Wolf SE, Tanzi RE, Kim DY. The Impact of the Cellular Environment and Aging on Modeling Alzheimer's Disease in 3D Cell Culture Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205037. [PMID: 36642841 PMCID: PMC10015857 DOI: 10.1002/advs.202205037] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/29/2022] [Indexed: 06/13/2023]
Abstract
Creating a cellular model of Alzheimer's disease (AD) that accurately recapitulates disease pathology has been a longstanding challenge. Recent studies showed that human AD neural cells, integrated into three-dimensional (3D) hydrogel matrix, display key features of AD neuropathology. Like in the human brain, the extracellular matrix (ECM) plays a critical role in determining the rate of neuropathogenesis in hydrogel-based 3D cellular models. Aging, the greatest risk factor for AD, significantly alters brain ECM properties. Therefore, it is important to understand how age-associated changes in ECM affect accumulation of pathogenic molecules, neuroinflammation, and neurodegeneration in AD patients and in vitro models. In this review, mechanistic hypotheses is presented to address the impact of the ECM properties and their changes with aging on AD and AD-related dementias. Altered ECM characteristics in aged brains, including matrix stiffness, pore size, and composition, will contribute to disease pathogenesis by modulating the accumulation, propagation, and spreading of pathogenic molecules of AD. Emerging hydrogel-based disease models with differing ECM properties provide an exciting opportunity to study the impact of brain ECM aging on AD pathogenesis, providing novel mechanistic insights. Understanding the role of ECM aging in AD pathogenesis should also improve modeling AD in 3D hydrogel systems.
Collapse
Affiliation(s)
- Matthias Hebisch
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Stefanie Klostermeier
- Institute of Medical PhysicsFriedrich‐Alexander Universität Erlangen‐Nürnberg91052ErlangenGermany
- Max‐Planck‐Zentrum für Physik und Medizin91054ErlangenGermany
| | - Katharina Wolf
- Department of Medicine 1Friedrich‐Alexander‐Universität Erlangen‐Nürnberg91054ErlangenGermany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg91058ErlangenGermany
| | - Stephan E. Wolf
- Institute of Glass and CeramicsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg91058ErlangenGermany
| | - Rudolph E. Tanzi
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Doo Yeon Kim
- Genetics and Aging Research UnitMcCance Center for Brain health, MassGeneral Institute for Neurodegenerative DiseaseMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| |
Collapse
|
8
|
Mor N, Raghav N. In-vitro simulation of modified-alginate ester as sustained release delivery system for curcumin. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
9
|
Comparative Study of Physicochemical Properties of Alginate Composite Hydrogels Prepared by the Physical Blending and Electrostatic Assembly Methods. Gels 2022; 8:gels8120799. [PMID: 36547323 PMCID: PMC9777933 DOI: 10.3390/gels8120799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Alginate hydrogel commonly suffers from defects, such as weak mechanical properties, the shortage of long-term stability in physiological medium and the lack of mammalian cell adhesivity due to its strong hydrophilicity in biomedical application. For this reason, the homogeneous alginate hydrogels (Alg Gel) were successfully prepared by the D-glucono-δ-lactone/hydroxyapatite (HAP/GDL) cross-linking system, and then, the physical blending and alternating electrostatic assembly technology were proposed to fabricate alginate composite hydrogels (Alg-GT, Alg-CS-GT and ALG/GT-CS). The feasibility of the design methods was verified through the comparative analysis of their physicochemical properties and biological activity. In particular, the effects of physical blending and alternating electrostatic assembly technology on the pore structure, mechanical properties, swelling, degradation, cell adhesion and proliferation of composite hydrogels were also investigated. Experimental results showed that the formation of polyelectrolyte complexes by electrostatic assembly between biological macromolecules and the covalent cross-linking of EDC/NHS to GT improved the vulnerability of ion cross-linking, enhanced the mechanical properties and swelling stability of the composite hydrogels, and regulated their pore structure and in vitro biodegradability properties. Furthermore, MC3T3-E1 cells could exhibit good cell adhesion, cell viability and cell proliferation on the alginate composite hydrogels. Among them, Alg-CS-GT showed the best cell proliferation ability and differentiation effect due to its good cell adhesion. In view of the excellent physicochemical properties and biological activity of Alg-CS-GT, it exhibited great potential in biomedical application for tissue engineering.
Collapse
|
10
|
Goncalves KE, Phillips S, Shah DSH, Athey D, Przyborski SA. Application of biomimetic surfaces and 3D culture technology to study the role of extracellular matrix interactions in neurite outgrowth and inhibition. BIOMATERIALS ADVANCES 2022; 144:213204. [PMID: 36434926 DOI: 10.1016/j.bioadv.2022.213204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
The microenvironment that cells experience during in vitro culture can often be far removed from the native environment they are exposed to in vivo. To recreate the physiological environment that developing neurites experience in vivo, we combine a well-established model of human neurite development with, functionalisation of both 2D and 3D growth substrates with specific extracellular matrix (ECM) derived motifs displayed on engineered scaffold proteins. Functionalisation of growth substrates provides biochemical signals more reminiscent of the in vivo environment and the combination of this technology with 3D cell culture techniques, further recapitulates the native cellular environment by providing a more physiologically relevant geometry for neurites to develop. This biomaterials approach was used to study interactions between the ECM and developing neurites, along with the identification of specific motifs able to enhance neuritogenesis within this model. Furthermore, this technology was employed to study the process of neurite inhibition that has a detrimental effect on neuronal connectivity following injury to the central nervous system (CNS). Growth substrates were functionalised with inhibitory peptides released from damaged myelin within the injured spinal cord (Nogo & OMgp). This model was then utilised to study the underlying molecular mechanisms that govern neurite inhibition in addition to potential mechanisms of recovery.
Collapse
Affiliation(s)
- K E Goncalves
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - S Phillips
- Orla Protein Technologies Ltd, (now part of Porvair Sciences Ltd), 73 Clywedog Road East, Wrexham Industrial Estate, Wrexham LL13 9XS, UK
| | - D S H Shah
- Orla Protein Technologies Ltd, (now part of Porvair Sciences Ltd), 73 Clywedog Road East, Wrexham Industrial Estate, Wrexham LL13 9XS, UK
| | - D Athey
- Orla Protein Technologies Ltd, (now part of Porvair Sciences Ltd), 73 Clywedog Road East, Wrexham Industrial Estate, Wrexham LL13 9XS, UK
| | - S A Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; Reprocell Europe Ltd, NETPark Incubator, Thomas Wright Way, Sedgefield TS21 3FD, UK.
| |
Collapse
|
11
|
Janzen D, Bakirci E, Faber J, Andrade Mier M, Hauptstein J, Pal A, Forster L, Hazur J, Boccaccini AR, Detsch R, Teßmar J, Budday S, Blunk T, Dalton PD, Villmann C. Reinforced Hyaluronic Acid-Based Matrices Promote 3D Neuronal Network Formation. Adv Healthc Mater 2022; 11:e2201826. [PMID: 35993391 PMCID: PMC11468248 DOI: 10.1002/adhm.202201826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Indexed: 01/28/2023]
Abstract
3D neuronal cultures attempt to better replicate the in vivo environment to study neurological/neurodegenerative diseases compared to 2D models. A challenge to establish 3D neuron culture models is the low elastic modulus (30-500 Pa) of the native brain. Here, an ultra-soft matrix based on thiolated hyaluronic acid (HA-SH) reinforced with a microfiber frame is formulated and used. Hyaluronic acid represents an essential component of the brain extracellular matrix (ECM). Box-shaped frames with a microfiber spacing of 200 µm composed of 10-layers of poly(ɛ-caprolactone) (PCL) microfibers (9.7 ± 0.2 µm) made via melt electrowriting (MEW) are used to reinforce the HA-SH matrix which has an elastic modulus of 95 Pa. The neuronal viability is low in pure HA-SH matrix, however, when astrocytes are pre-seeded below this reinforced construct, they significantly support neuronal survival, network formation quantified by neurite length, and neuronal firing shown by Ca2+ imaging. The astrocyte-seeded HA-SH matrix is able to match the neuronal viability to the level of Matrigel, a gold standard matrix for neuronal culture for over two decades. Thus, this 3D MEW frame reinforced HA-SH composite with neurons and astrocytes constitutes a reliable and reproducible system to further study brain diseases.
Collapse
Affiliation(s)
- Dieter Janzen
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Ezgi Bakirci
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Jessica Faber
- Department of Mechanical EngineeringInstitute of Applied MechanicsFriedrich‐Alexander University of Erlangen‐NürnbergEgerlandstrasse 591058ErlangenGermany
| | - Mateo Andrade Mier
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Julia Hauptstein
- Department of TraumaHand, Plastic and Reconstructive SurgeryUniversity Hospital WürzburgOberdürrbacher Str. 697080WürzburgGermany
| | - Arindam Pal
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Leonard Forster
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
| | - Jonas Hazur
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander University of Erlangen‐NürnbergCauerstr. 691058ErlangenGermany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander University of Erlangen‐NürnbergCauerstr. 691058ErlangenGermany
| | - Rainer Detsch
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander University of Erlangen‐NürnbergCauerstr. 691058ErlangenGermany
| | - Jörg Teßmar
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
| | - Silvia Budday
- Department of Mechanical EngineeringInstitute of Applied MechanicsFriedrich‐Alexander University of Erlangen‐NürnbergEgerlandstrasse 591058ErlangenGermany
| | - Torsten Blunk
- Department of TraumaHand, Plastic and Reconstructive SurgeryUniversity Hospital WürzburgOberdürrbacher Str. 697080WürzburgGermany
| | - Paul D. Dalton
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
- Phil and Penny Knight Campus for Accelerating Scientific ImpactUniversity of Oregon1505 Franklin BlvdEugeneOR97403USA
| | - Carmen Villmann
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| |
Collapse
|
12
|
Tyramine-Functionalized Alginate-Collagen Hybrid Hydrogel Inks for 3D-Bioprinting. Polymers (Basel) 2022; 14:polym14153173. [PMID: 35956690 PMCID: PMC9371113 DOI: 10.3390/polym14153173] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/02/2022] Open
Abstract
Extrusion-based 3D-bioprinting using hydrogels has exhibited potential in precision medicine; however, researchers are beset with several challenges. A major challenge of this technique is the production of constructs with sufficient height and fidelity to support cellular behavior in vivo. In this study, we present the 3D-bioprinting of cylindrical constructs with tunable gelation kinetics by controlling the covalent crosslinking density and gelation time of a tyramine-functionalized alginate hydrogel (ALG-TYR) via enzymatic reaction by horseradish peroxidase (HRP) and hydrogen peroxide (H2O2). The extruded filament was crosslinked for a second time on a support bath containing H2O2 to increase fidelity after printing. The resulting tubular construct, with a height of 6 mm and a wall thickness of 2 mm, retained its mechanical properties and had a maximum 2-fold swelling after 2 d. Furthermore, collagen (COL) was introduced into the ALG-TYR hydrogel network to increase the mechanical modulus and cell cytocompatibility, as the encapsulated fibroblast cells exhibited a higher cell viability in the ALG-TYR/COL construct (92.13 ± 0.70%) than in ALG-TYR alone (68.18 ± 3.73%). In summary, a vascular ECM-mimicking scaffold was 3D-bioprinted with the ALG-TYR/COL hybrid hydrogel, and this scaffold can support tissue growth for clinical translation in regenerative and personalized medicine.
Collapse
|
13
|
Jarrah R, Sammak SE, Onyedimma C, Ghaith AK, Moinuddin F, Bhandarkar AR, Siddiqui A, Madigan N, Bydon M. The Role of Alginate Hydrogels as a Potential Treatment Modality for Spinal Cord Injury: A Comprehensive Review of the Literature. Neurospine 2022; 19:272-280. [PMID: 35793929 PMCID: PMC9260541 DOI: 10.14245/ns.2244186.093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/23/2022] [Indexed: 12/14/2022] Open
Abstract
Objective To comprehensively characterize the utilization of alginate hydrogels as an alternative treatment modality for spinal cord injury (SCI).
Methods An extensive review of the published literature on studies using alginate hydrogels to treat SCI was performed. The review of the literature was performed using electronic databases such as PubMed, EMBASE, and OVID MEDLINE electronic databases. The keywords used were “alginate,” “spinal cord injury,” “biomaterial,” and “hydrogel.”
Results In the literature, we identified a total of 555 rat models that were treated with alginate scaffolds for regenerative biomarkers. Alginate hydrogels were found to be efficient and promising substrates for tissue engineering, drug delivery, neural regeneration, and cellbased therapies for SCI repair. With its ability to act as a pro-regenerative and antidegenerative agent, the alginate hydrogel has the potential to improve clinical outcomes.
Conclusion The emerging developments of alginate hydrogels as treatment modalities may support current and future tissue regenerative strategies for SCI.
Collapse
Affiliation(s)
- Ryan Jarrah
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Sally El Sammak
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Chiduziem Onyedimma
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Abdul Karim Ghaith
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - F.M. Moinuddin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Archis R. Bhandarkar
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Ahad Siddiqui
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Mohamad Bydon
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
- Corresponding Author Mohamad Bydon Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, Minnesota, USA
| |
Collapse
|
14
|
Mabrouk M, Ismail E, Beherei H, Abo-Elfadl MT, Salem ZA, Das DB, AbuBakr N. Biocompatibility of hydroxyethyl cellulose/glycine/RuO 2 composite scaffolds for neural-like cells. Int J Biol Macromol 2022; 209:2097-2108. [PMID: 35504415 DOI: 10.1016/j.ijbiomac.2022.04.190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/24/2022]
Abstract
Fabrication of scaffolds for nerve regeneration is one of the most challenging topics in regenerative medicine at the moment, which is also interlinked with the development of biocompatible substrates for cells growth. This work is targeted towards the development of green biomaterial composite scaffolds for nerve cell culture applications. Hybrid scaffolds of hydroxyethyl cellulose/glycine (HEC/Gly) composite doped with different concentrations of green ruthenium oxide (RuO2) were synthesized and characterized via a combination of different techniques. X-rays diffraction (XRD) and differential scanning calorimetry (DSC) analyses showed a crystalline nature for all the samples with noticeable decrease in the peak intensity of the fabricated scaffolds as compared to that for pure glycine. Fourier transform infrared spectroscopy (FTIR) tests revealed an increase in the vibrational bands of the synthesized RuO2 containing scaffolds which are related to the functional groups of the natural plant extract (Aspalathuslinearis) used for RuO2 nanoparticles (NPs) synthesis. Scanning electron microscopy (SEM) results revealed a 3D porous structure of the scaffolds with variant features attributed to the concentration of RuO2 NPs in the scaffold. The compressive test results recorded an enhancement in mechanical properties of the fabricated scaffolds (up to 8.55 MPa), proportionally correlated to increasing the RuO2 NPs concentration in HEC/Gly composite scaffold. Our biocompatibility tests revealed that the composite scaffolds doped with 1 and 2 ml of RuO2 demonstrated the highest proliferation percentages (152.2 and 135.6%) compared to control. Finally, the SEM analyses confirmed the impressive cells attachments and differentiation onto the scaffold surfaces as evidenced by the presence of many neuron-like cells with apparent cell bodies and possessing few short neurite-like processes. The presence of RuO2 and glycine was due to their extraordinary biocompatibility due to their cytoprotective and regenerative effects. Therefore, we conclude that these scaffolds are promising for accommodation and growth of neural-like cells.
Collapse
Affiliation(s)
- Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33El Bohouth St. (former EL Tahrir St.), Dokki, Giza, P.O.12622, Egypt.
| | - Enas Ismail
- Department of Restorative Dentistry, Faculty of Dentistry, University of the Western Cape, Cape Town 7505, South Africa; Physics Department, Faculty of Science (Girl's branch), Al Azhar University, Nasr City, Cairo, Egypt
| | - Hanan Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33El Bohouth St. (former EL Tahrir St.), Dokki, Giza, P.O.12622, Egypt.
| | - Mahmoud T Abo-Elfadl
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, 33 El-Buhouth Street, Dokki, Giza 12622, Egypt; Biochemistry Department, National Research Centre, Dokki, Giza, Egypt
| | - Zeinab A Salem
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo, Egypt; Faculty of Oral and Dental Medicine, Ahram Canadian University, Cairo, Egypt
| | - Diganta B Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, Leicestershire, UK
| | - Nermeen AbuBakr
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo, Egypt; Stem Cells and Tissue Engineering Unit, Faculty of Dentistry, Cairo University, Cairo, Egypt
| |
Collapse
|
15
|
Hegab R, Van Volkenburg T, Ohiri K, Sebeck N, Bessling S, Theodore M, Rossick K, Pellicore M, Benkoski J, Patrone J. Design of experiments approach to developing a robust ink for bioprinting. Biomed Phys Eng Express 2022; 8. [PMID: 35290975 DOI: 10.1088/2057-1976/ac5de1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/15/2022] [Indexed: 11/11/2022]
Abstract
Despite advancements in tissue engineering, the methods used to generate three-dimensional (3D)in vitromodels for rapid screening and characterization studies remain time and labor intensive. Bioprinting offers an opportunity to offset these limitations by providing a scalable, high-throughput method with precise control over biomaterial scaffold and cellular deposition. However, the process of formulating bioinks can be complex in terms of balancing the mechanical integrity of a bioscaffold and viability of cells. One key factor, especially in alginate-based bioinks, is the rate of bioscaffold dissolution. It must allow cells to replace the bioscaffold with extracellular matrix (ECM), yet remain durable during extended tissue culture. This study uses a Design of Experiments (DoE) approach to understand the dependencies of multiple variables involved in the formulation and processing of an alginate-based bioink. The focus of the DoE was to understand the effects of hydrogel composition on bioink durability while maintaining cell viability. Three ingredients were varied in all: alginate, nanocellulose, and fibrinogen. Their effects on the bioink were then measured with respect to extrudability, strength, and stiffness as determined by dynamic mechanical analysis (DMA). The DoE demonstrated that mechanical integrity increased with increasing alginate concentration. In contrast, fibrinogen and nanofibril concentration had no statistically significant effect. The optimized ink containing fibroblasts was printable using multiple nozzle sizes while also supporting fibroblast cell viability. DMA characterization further showed that the composition of the cell culture medium did not modulate the degradation rate of the hydrogel. Ultimately, the study outlines a methodology for formulating a bioink that will result in robust bioscaffolds forin vitromodel development.
Collapse
Affiliation(s)
- Rachel Hegab
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Tessa Van Volkenburg
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Korine Ohiri
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Natalie Sebeck
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Seneca Bessling
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Mellisa Theodore
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Katelyn Rossick
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Matthew Pellicore
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Jason Benkoski
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| | - Julia Patrone
- The Johns Hopkins Applied Physics Laboratory, Laurel, MD, United States of America
| |
Collapse
|
16
|
Mohan T, Kleinschek KS, Kargl R. Polysaccharide peptide conjugates: Chemistry, properties and applications. Carbohydr Polym 2022; 280:118875. [PMID: 35027118 DOI: 10.1016/j.carbpol.2021.118875] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 11/02/2022]
Abstract
The intention of this publication is to give an overview on research related to conjugates of polysaccharides and peptides. Dextran, chitosan, and alginate were selected, to cover four of the most often encountered functional groups known to be present in polysaccharides. These groups are the hydroxyl, the amine, the carboxyl, and the acetal functionality. A collection of the commonly used chemical reactions for conjugation is provided. Conjugation results into distinct properties compared to the parent polysaccharide, and a number of these characteristics are highlighted. This review aims at demonstrating the applicability of said conjugates with a strong emphasis on biomedical applications, drug delivery, biosensing, and tissue engineering. Some suggestions are made for more rigorous chemistries and analytics that could be investigated. Finally, an outlook is given into which direction the field could be developed further. We hope that this survey provides the reader with a comprehensive summary and contributes to the progress of works that aim at synthetically combining two of the main building blocks of life into supramolecular structures with unprecedented biological response.
Collapse
Affiliation(s)
- Tamilselvan Mohan
- Institute for Chemistry and Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Karin Stana Kleinschek
- Institute for Chemistry and Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Rupert Kargl
- Institute for Chemistry and Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria; Institute for Automation, Faculty of Electrical Engineering and Computer Science, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia.
| |
Collapse
|
17
|
Eleftheriadou D, Evans RE, Atkinson E, Abdalla A, Gavins FKH, Boyd AS, Williams GR, Knowles JC, Roberton VH, Phillips JB. An alginate-based encapsulation system for delivery of therapeutic cells to the CNS. RSC Adv 2022; 12:4005-4015. [PMID: 35425456 PMCID: PMC8981497 DOI: 10.1039/d1ra08563h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/22/2022] [Indexed: 12/21/2022] Open
Abstract
Treatment options for neurodegenerative conditions such as Parkinson's disease have included the delivery of cells which release dopamine or neurotrophic factors to the brain. Here, we report the development of a novel approach for protecting cells after implantation into the central nervous system (CNS), by developing dual-layer alginate beads that encapsulate therapeutic cells and release an immunomodulatory compound in a sustained manner. An optimal alginate formulation was selected with a view to providing a sustained physical barrier between engrafted cells and host tissue, enabling exchange of small molecules while blocking components of the host immune response. In addition, a potent immunosuppressant, FK506, was incorporated into the outer layer of alginate beads using electrosprayed poly-ε-caprolactone core–shell nanoparticles with prolonged release profiles. The stiffness, porosity, stability and ability of the alginate beads to support and protect encapsulated SH-SY5Y cells was demonstrated, and the release profile of FK506 and its effect on T-cell proliferation in vitro was characterized. Collectively, our results indicate this multi-layer encapsulation technology has the potential to be suitable for use in CNS cell delivery, to protect implanted cells from host immune responses whilst providing permeability to nutrients and released therapeutic molecules. Novel composite cell encapsulation system: dual-layer, micro-scale beads maintain cell survival while releasing immunomodulatory FK506 in a sustained manner. This biotechnology platform could be applicable for treatment of CNS and other disorders.![]()
Collapse
Affiliation(s)
- Despoina Eleftheriadou
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Rachael E Evans
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Emily Atkinson
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Ahmed Abdalla
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Francesca K H Gavins
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Ashleigh S Boyd
- UCL Institute of Immunity and Transplantation, Royal Free Hospital London UK
| | - Gareth R Williams
- UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Jonathan C Knowles
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute London UK
| | - Victoria H Roberton
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - James B Phillips
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| |
Collapse
|
18
|
Bonany M, del-Mazo-Barbara L, Espanol M, Ginebra MP. Microsphere incorporation as a strategy to tune the biological performance of bioinks. J Tissue Eng 2022; 13:20417314221119895. [PMID: 36199978 PMCID: PMC9527984 DOI: 10.1177/20417314221119895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Although alginate is widely used as a matrix in the formulation of cell-laden inks, this polymer often requires laborious processing strategies due to its lack of cell adhesion moieties. The main objective of the present work was to explore the incorporation of microspheres into alginate-based bioinks as a simple and tuneable way to solve the cell adhesion problems, while adding extra biological functionality and improving their mechanical properties. To this end, three types of microspheres with different mineral contents (i.e. gelatine with 0% of hydroxyapatite, gelatine with 25 wt% of hydroxyapatite nanoparticles and 100 wt% of calcium -deficient hydroxyapatite) were synthesised and incorporated into the formulation of cell-laden inks. The results showed that the addition of microspheres generally improved the rheological properties of the ink, favoured cell proliferation and positively affected osteogenic cell differentiation. Furthermore, this differentiation was found to be influenced by the type of microsphere and the ability of the cells to migrate towards them, which was highly dependent on the stiffness of the bioink. In this regard, Ca2+ supplementation in the cell culture medium had a pronounced effect on the relaxation of the stiffness of these cell-loaded inks, influencing the overall cell performance. In conclusion, we have developed a powerful and tuneable strategy for the fabrication of alginate-based bioinks with enhanced biological characteristics by incorporating microspheres into the initial ink formulation.
Collapse
Affiliation(s)
- Mar Bonany
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
| | - Laura del-Mazo-Barbara
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
| | - Montserrat Espanol
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
19
|
Dumbleton J, Shamul JG, Jiang B, Agarwal P, Huang H, Jia X, He X. Oxidation and RGD Modification Affect the Early Neural Differentiation of Murine Embryonic Stem Cells Cultured in Core-Shell Alginate Hydrogel Microcapsules. Cells Tissues Organs 2022; 211:294-303. [PMID: 34038907 PMCID: PMC8617071 DOI: 10.1159/000514580] [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: 10/21/2020] [Accepted: 01/11/2021] [Indexed: 01/03/2023] Open
Abstract
Directed neural differentiation of embryonic stem cells (ESCs) has been studied extensively to improve the treatment of neurodegenerative disorders. This can be done through stromal-cell derived inducing activity (SDIA), by culturing ESCs directly on top of a layer of feeder stromal cells. However, the stem cells usually become mixed with the feeder cells during the differentiation process, making it difficult to obtain a pure population of the differentiated cells for further use. To address this issue, a non-planar microfluidic device is used here to encapsulate murine ESCs (mESCs) in the 3D liquid core of microcapsules with an alginate hydrogel shell of different sizes for early neural differentiation through SDIA, by culturing mESC-laden microcapsules over a feeder layer of PA6 cells. Furthermore, the alginate hydrogel shell of the microcapsules is modified via oxidation or RGD peptide conjugation to examine the mechanical and chemical effects on neural differentiation of the encapsulated mESC aggregates. A higher expression of Nestin is observed in the aggregates encapsulated in small (∼300 μm) microcapsules and cultured over the PA6 cell feeder layer. Furthermore, the modification of the alginate with RGD facilitates early neurite extension within the microcapsules. This study demonstrates that the presence of the RGD peptide, the SDIA effect of the PA6 cells, and the absence of leukemia inhibition factor from the medium can lead to the early differentiation of mESCs with extensive neurites within the 3D microenvironment of the small microcapsules. This is the first study to investigate the effects of cell adhesion and degradation of the encapsulation materials for directed neural differentiation of mESCs. The simple modifications (i.e., oxidation and RGD incorporation) of the miniaturized 3D environment for improved early neural differentiation of mESCs may potentially enhance further downstream differentiation of the mESCs into more specialized neurons for therapeutic use and drug screening.
Collapse
Affiliation(s)
- Jenna Dumbleton
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA)
| | - James G. Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Bin Jiang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Pranay Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA)
| | - Haishui Huang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA)
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA),Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA,Correspondence should be addressed to: Xiaoming He, Ph.D., Fishell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, Phone: 301-405-7946,
| |
Collapse
|
20
|
Ladeira B, Custodio C, Mano J. Core-Shell Microcapsules: Biofabrication and Potential Applications in Tissue Engineering and Regenerative Medicine. Biomater Sci 2022; 10:2122-2153. [DOI: 10.1039/d1bm01974k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The construction of biomaterial scaffolds that accurately recreate the architecture of living tissues in vitro is a major challenge in the field of tissue engineering and regenerative medicine. Core-shell microcapsules...
Collapse
|
21
|
Rosiak P, Latanska I, Paul P, Sujka W, Kolesinska B. Modification of Alginates to Modulate Their Physic-Chemical Properties and Obtain Biomaterials with Different Functional Properties. Molecules 2021; 26:7264. [PMID: 34885846 PMCID: PMC8659150 DOI: 10.3390/molecules26237264] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
Modified alginates have a wide range of applications, including in the manufacture of dressings and scaffolds used for regenerative medicine, in systems for selective drug delivery, and as hydrogel materials. This literature review discusses the methods used to modify alginates and obtain materials with new or improved functional properties. It discusses the diverse biological and functional activity of alginates. It presents methods of modification that utilize both natural and synthetic peptides, and describes their influence on the biological properties of the alginates. The success of functionalization depends on the reaction conditions being sufficient to guarantee the desired transformations and provide modified alginates with new desirable properties, but mild enough to prevent degradation of the alginates. This review is a literature description of efficient methods of alginate functionalization using biologically active ligands. Particular attention was paid to methods of alginate functionalization with peptides, because the combination of the properties of alginates and peptides leads to the obtaining of conjugates with properties resulting from both components as well as a completely new, different functionality.
Collapse
Affiliation(s)
- Piotr Rosiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Ilona Latanska
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Paulina Paul
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Witold Sujka
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| |
Collapse
|
22
|
Hydrogel, Electrospun and Composite Materials for Bone/Cartilage and Neural Tissue Engineering. MATERIALS 2021; 14:ma14226899. [PMID: 34832300 PMCID: PMC8624846 DOI: 10.3390/ma14226899] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Injuries of the bone/cartilage and central nervous system are still a serious socio-economic problem. They are an effect of diversified, difficult-to-access tissue structures as well as complex regeneration mechanisms. Currently, commercially available materials partially solve this problem, but they do not fulfill all of the bone/cartilage and neural tissue engineering requirements such as mechanical properties, biochemical cues or adequate biodegradation. There are still many things to do to provide complete restoration of injured tissues. Recent reports in bone/cartilage and neural tissue engineering give high hopes in designing scaffolds for complete tissue regeneration. This review thoroughly discusses the advantages and disadvantages of currently available commercial scaffolds and sheds new light on the designing of novel polymeric scaffolds composed of hydrogels, electrospun nanofibers, or hydrogels loaded with nano-additives.
Collapse
|
23
|
Mrówczyńska E, Mazur AJ. Integrin-Linked Kinase (ILK) Plays an Important Role in the Laminin-Dependent Development of Dorsal Root Ganglia during Chicken Embryogenesis. Cells 2021; 10:cells10071666. [PMID: 34359835 PMCID: PMC8304069 DOI: 10.3390/cells10071666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022] Open
Abstract
Integrin-linked kinase (ILK) is mainly localized in focal adhesions where it interacts and modulates the downstream signaling of integrins affecting cell migration, adhesion, and survival. The interaction of dorsal root ganglia (DRG) cells, being part of the peripheral nervous system (PNS), with the extracellular matrix (ECM) via integrins is crucial for proper PNS development. A few studies have focused on ILK’s role in PNS development, but none of these have focused on chicken. Therefore, we decided to investigate ILK’s role in the development of Gallus gallus domesticus’s DRG. First, using RT-PCR, Western blotting, and in situ hybridization, we show that ILK is expressed in DRG. Next, by immunocytochemistry, we show ILK’s localization both intracellularly and on the cell membrane of DRG neurons and Schwann cell precursors (SCPs). Finally, we describe ILK’s involvement in multiple aspects of DRG development by performing functional experiments in vitro. IgG-mediated interruption of ILK’s action improved DRG neurite outgrowth, modulated their directionality, stimulated SCPs migration, and impacted growth cone morphology in the presence of laminin-1 or laminin-1 mimicking peptide IKVAV. Taken together, our results show that ILK is important for chicken PNS development, probably via its exposure to the ECM.
Collapse
Affiliation(s)
- Ewa Mrówczyńska
- Correspondence: (E.M.); (A.J.M.); Tel.: +48-71-375-7972 (E.M.); +48-71-375-6206 (A.J.M.)
| | - Antonina Joanna Mazur
- Correspondence: (E.M.); (A.J.M.); Tel.: +48-71-375-7972 (E.M.); +48-71-375-6206 (A.J.M.)
| |
Collapse
|
24
|
Maruyama M, Moeinzadeh S, Guzman RA, Zhang N, Storaci HW, Utsunomiya T, Lui E, Huang EE, Rhee C, Gao Q, Yao Z, Takagi M, Yang YP, Goodman SB. The efficacy of lapine preconditioned or genetically modified IL4 over-expressing bone marrow-derived mesenchymal stromal cells in corticosteroid-associated osteonecrosis of the femoral head in rabbits. Biomaterials 2021; 275:120972. [PMID: 34186237 DOI: 10.1016/j.biomaterials.2021.120972] [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: 12/04/2020] [Revised: 05/29/2021] [Accepted: 06/10/2021] [Indexed: 02/09/2023]
Abstract
Cell-based therapy for augmentation of core decompression (CD) using mesenchymal stromal cells (MSCs) is a promising treatment for early stage osteonecrosis of the femoral head (ONFH). Recently, the therapeutic potential for immunomodulation of osteogenesis using preconditioned (with pro-inflammatory cytokines) MSCs (pMSCs), or by the timely resolution of inflammation using MSCs that over-express anti-inflammatory cytokines has been described. Here, pMSCs exposed to tumor necrosis factor-alpha and lipopolysaccharide for 3 days accelerated osteogenic differentiation in vitro. Furthermore, injection of pMSCs encapsulated with injectable hydrogels into the bone tunnel facilitated angiogenesis and osteogenesis in the femoral head in vivo, using rabbit bone marrow-derived MSCs and a model of corticosteroid-associated ONFH in rabbits. In contrast, in vitro and in vivo studies demonstrated that genetically-modified MSCs that over-express IL4 (IL4-MSCs), established by using a lentiviral vector carrying the rabbit IL4 gene under the cytomegalovirus promoter, accelerated proliferation of MSCs and decreased the percentage of empty lacunae in the femoral head. Therefore, adjunctive cell-based therapy of CD using pMSCs and IL4-MSCs may hold promise to heal osteonecrotic lesions in the early stage ONFH. These interventions must be applied in a temporally sensitive fashion, without interfering with the mandatory acute inflammatory phase of bone healing.
Collapse
Affiliation(s)
- Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberto Alfonso Guzman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hunter W Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Mechanical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA; Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
25
|
Marinval N, Chew SY. Mechanotransduction assays for neural regeneration strategies: A focus on glial cells. APL Bioeng 2021; 5:021505. [PMID: 33948526 PMCID: PMC8088332 DOI: 10.1063/5.0037814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/19/2021] [Indexed: 01/22/2023] Open
Abstract
Glial cells are mechanosensitive, and thus, engineered systems have taken a step forward to design mechanotransduction platforms in order to impart diverse mechanical stresses to cells. Mechanical strain encountered in the central nervous system can arise from diverse mechanisms, such as tissue reorganization, fluid flow, and axon growth, as well as pathological events including axon swelling or mechanical trauma. Biomechanical relevance of the in vitro mechanical testing requires to be placed in line with the physiological and mechanical changes in central nervous tissues that occur during the progression of neurodegenerative diseases. Mechanotransduction signaling utilized by glial cells and the recent approaches intended to model altered microenvironment adapted to pathological context are discussed in this review. New insights in systems merging substrate's stiffness and topography should be considered for further glial mechanotransduction studies, while testing platforms for drug discoveries promise great advancements in pharmacotherapy. Potential leads and strategies for clinical outcomes are expected to be developed following the exploration of these glial mechanosensitive signaling pathways.
Collapse
Affiliation(s)
- Nicolas Marinval
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - Sing Yian Chew
- Author to whom correspondence should be addressed: . Tel.: +65 6316 8812. Fax: +65 6794 7553
| |
Collapse
|
26
|
Saghati S, Rahbarghazi R, Baradar Khoshfetrat A, Moharamzadeh K, Tayefi Nasrabadi H, Roshangar L. Phenolated alginate-collagen hydrogel induced chondrogenic capacity of human amniotic mesenchymal stem cells. J Biomater Appl 2021; 36:789-802. [PMID: 34074175 DOI: 10.1177/08853282211021692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Horseradish peroxidase (HRP)-catalyzed hydrogels are considered to be an important platform for tissue engineering applications. In this study, we investigated the chondrogenic capacity of phenolated (1.2%) alginate-(0.5%) collagen hydrogel on human amniotic mesenchymal stem cells after 21 days. Using NMR, FTIR analyses, and SEM imaging, we studied the phenolation and structure of alginate-collagen hydrogel. For physicochemical evaluations, gelation time, mechanical properties, swelling, and degradation rate were assessed. The survival rate was monitored using the MTT assay and DAPI staining. Western blotting was performed to measure the chondrogenic differentiation of cells. NMR showed successful phenolation of the alginate-collagen hydrogel. FTIR exhibited the interaction between the functional groups of collagen with phenolated alginate. SEM showed the existence of collagen microfibrils in the alginate-collagen hydrogel. Compared to phenolated alginate, the addition of collagen increased hydrogel elasticity by 10%. Both swelling rate and biodegradability were reduced in the presence of collagen. We noted an increased survival rate in phenolated alginate-collagen compared to the control cells (p < 0.05). Western blotting revealed the increase of chondrocyte-associated proteins such as SOX9 and COL2A1 in phenolated-alginate-collagen hydrogels after 21 days. These data showed that phenolated alginate-collagen hydrogel is an appropriate 3 D substrate to induce chondrogenic capacity of human mesenchymal stem cells.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Baradar Khoshfetrat
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamid Tayefi Nasrabadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Lu L, Duong VT, Shalash AO, Skwarczynski M, Toth I. Chemical Conjugation Strategies for the Development of Protein-Based Subunit Nanovaccines. Vaccines (Basel) 2021; 9:563. [PMID: 34071482 PMCID: PMC8228360 DOI: 10.3390/vaccines9060563] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
The production of subunit nanovaccines relies heavily on the development of a vaccine delivery system that is safe and efficient at delivering antigens to the target site. Nanoparticles have been extensively investigated for vaccine delivery over the years, as they often possess self-adjuvanting properties. The conjugation of antigens to nanoparticles by covalent bonds ensures co-delivery of these components to the same subset of immune cells in order to trigger the desired immune responses. Herein, we review covalent conjugation strategies for grafting protein or peptide antigens onto other molecules or nanoparticles to obtain subunit nanovaccines. We also discuss the advantages of chemical conjugation in developing these vaccines.
Collapse
Affiliation(s)
| | | | | | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (L.L.); (V.T.D.); (A.O.S.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (L.L.); (V.T.D.); (A.O.S.)
| |
Collapse
|
28
|
Golunova A, Velychkivska N, Mikšovská Z, Chochola V, Jaroš J, Hampl A, Pop-Georgievski O, Proks V. Direct and Indirect Biomimetic Peptide Modification of Alginate: Efficiency, Side Reactions, and Cell Response. Int J Mol Sci 2021; 22:5731. [PMID: 34072085 PMCID: PMC8198284 DOI: 10.3390/ijms22115731] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022] Open
Abstract
In the fast-developing field of tissue engineering there is a constant demand for new materials as scaffolds for cell seeding, which can better mimic a natural extracellular matrix as well as control cell behavior. Among other materials, polysaccharides are widely used for this purpose. One of the main candidates for scaffold fabrication is alginate. However, it lacks sites for cell adhesion. That is why one of the steps toward the development of suitable scaffolds for cells is the introduction of the biofunctionality to the alginate structure. In this work we focused on bone-sialoprotein derived peptide (TYRAY) conjugation to the molecule of alginate. Here the comparison study on four different approaches of peptide conjugation was performed including traditional and novel modification methods, based on 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide/N-hydroxy succinimide (EDC/NHS), 4-(4,6-dimethoxy-1,3,5-triazine-2-yl)-4-methylmorpholinium chloride (DMTMM), thiol-Michael addition and Cu-catalyzed azide-alkyne cycloaddition reactions. It was shown that the combination of the alginate amidation with the use of and subsequent Cu-catalyzed azide-alkyne cycloaddition led to efficient peptide conjugation, which was proven with both NMR and XPS methods. Moreover, the cell culture experiment proved the positive effect of peptide presence on the adhesion of human embryonic stem cells.
Collapse
Affiliation(s)
- Anna Golunova
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky Sq. 2, 16206 Prague, Czech Republic; (N.V.); (Z.M.); (O.P.-G.); (V.P.)
| | - Nadiia Velychkivska
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky Sq. 2, 16206 Prague, Czech Republic; (N.V.); (Z.M.); (O.P.-G.); (V.P.)
| | - Zuzana Mikšovská
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky Sq. 2, 16206 Prague, Czech Republic; (N.V.); (Z.M.); (O.P.-G.); (V.P.)
| | - Václav Chochola
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czech Republic; (V.C.); (J.J.); (A.H.)
- Cell and Tissue Regeneration, International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 65691 Brno, Czech Republic
| | - Josef Jaroš
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czech Republic; (V.C.); (J.J.); (A.H.)
- Cell and Tissue Regeneration, International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 65691 Brno, Czech Republic
| | - Aleš Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czech Republic; (V.C.); (J.J.); (A.H.)
- Cell and Tissue Regeneration, International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 65691 Brno, Czech Republic
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky Sq. 2, 16206 Prague, Czech Republic; (N.V.); (Z.M.); (O.P.-G.); (V.P.)
| | - Vladimír Proks
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky Sq. 2, 16206 Prague, Czech Republic; (N.V.); (Z.M.); (O.P.-G.); (V.P.)
| |
Collapse
|
29
|
Carvalho IC, Mansur HS, Leonel AG, Mansur AAP, Lobato ZIP. Soft matter polysaccharide-based hydrogels as versatile bioengineered platforms for brain tissue repair and regeneration. Int J Biol Macromol 2021; 182:1091-1111. [PMID: 33892028 DOI: 10.1016/j.ijbiomac.2021.04.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023]
Abstract
Acute or chronic brain injuries promote deaths and the life-long debilitating neurological status where, despite advances in therapeutic strategies, clinical outcome hardly achieves total patient recovery. In recent decades, brain tissue engineering emerged as an encouraging area of research for helping in damaged central nervous system (CNS) recovery. Polysaccharides are abundant naturally occurring biomacromolecules with a great potential enhancement of advanced technologies in brain tissue repair and regeneration (BTRR). Besides carrying rich biological information, polysaccharides can interact and communicate with biomolecules, including glycosaminoglycans present in cell membranes and many signaling moieties, growth factors, chemokines, and axon guidance molecules. This review includes a comprehensive investigation of the current progress on designing and developing polysaccharide-based soft matter biomaterials for BTRR. Although few interesting reviews concerning BTRR have been reported, this is the first report specifically focusing on covering multiple polysaccharides and polysaccharide-based functionalized biomacromolecules in this emerging and intriguing field of multidisciplinary knowledge. This review aims to cover the state of art challenges and prospects of this fascinating field while presenting the richness of possibilities of using these natural biomacromolecules for advanced biomaterials in prospective neural tissue engineering applications.
Collapse
Affiliation(s)
- Isadora C Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Herman S Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil.
| | - Alice G Leonel
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Alexandra A P Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Zelia I P Lobato
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais - UFMG, Brazil
| |
Collapse
|
30
|
Teng K, An Q, Chen Y, Zhang Y, Zhao Y. Recent Development of Alginate-Based Materials and Their Versatile Functions in Biomedicine, Flexible Electronics, and Environmental Uses. ACS Biomater Sci Eng 2021; 7:1302-1337. [PMID: 33764038 DOI: 10.1021/acsbiomaterials.1c00116] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alginate is a natural polysaccharide that is easily chemically modified or compounded with other components for various types of functionalities. The alginate derivatives are appealing not only because they are biocompatible so that they can be used in biomedicine or tissue engineering but also because of the prospering bioelectronics that require various biomaterials to interface between human tissues and electronics or to serve as electronic components themselves. The study of alginate-based materials, especially hydrogels, have repeatedly found new frontiers over recent years. In this Review, we document the basic properties of alginate, their chemical modification strategies, and the recent development of alginate-based functional composite materials. The newly thrived functions such as ionically conductive hydrogel or 3D or 4D cell culturing matrix are emphasized among other appealing potential applications. We expect that the documentation of relevant information will stimulate scientific efforts to further develop biocompatible electronics or smart materials and to help the research domain better address the medicine, energy, and environmental challenges faced by human societies.
Collapse
Affiliation(s)
- Kaixuan Teng
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| | - Qi An
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| | - Yao Chen
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| | - Yihe Zhang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| | - Yantao Zhao
- Institute of Orthopedics, Fourth Medical Center of the General Hospital of CPLA, Beijing 100048, China.,Beijing Engineering Research Center of Orthopedics Implants, Beijing 100048, China
| |
Collapse
|
31
|
Moeinzadeh S, Park Y, Lin S, Yang YP. In-situ stable injectable collagen-based hydrogels for cell and growth factor delivery. MATERIALIA 2021; 15:100954. [PMID: 33367226 PMCID: PMC7751945 DOI: 10.1016/j.mtla.2020.100954] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Here we report development of in-situ stable injectable hydrogels for delivery of cells and growth factors based on two precursors, alginate, and collagen/calcium sulfate (CaSO4). The alg/col hydrogels were shear-thinning, injectable through commercially available needles and stable right after injection. Rheological measurements revealed that pre-crosslinked alg/col hydrogels fully crosslinked at 37°C and that the storage modulus of alg/col hydrogels increased with increasing the collagen content or the concentration of CaSO4. The viscoelastic characteristics and injectability of the alg/col hydrogels were not significantly impacted by the storage of precursor solutions for 28 days. An osteoinductive bone morphogenic protein-2 (BMP-2) loaded into alg/col hydrogels was released in 14 days. Human mesenchymal stem cells (hMSCs) encapsulated in alg/col hydrogels had over 90% viability over 7 days after injection. The DNA content of hMSC-laden alg/col hydrogels increased by 6-37 folds for 28 days, depending on the initial cell density. In addition, hMSCs encapsulated in alg/col hydrogels and incubated in osteogenic medium were osteogenically differentiated and formed a mineralized matrix. Finally, a BMP-2 loaded alg/col hydrogel was used to heal a critical size calvarial bone defect in rats after 8 weeks of injection. The alg/col hydrogel holds great promise in tissue engineering and bioprinting applications.
Collapse
Affiliation(s)
- Seyedsina Moeinzadeh
- Department of Orthopedic Surgery, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Youngbum Park
- Department of Orthopedic Surgery, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
- Department of Prosthodontics, Yonsei University College of Dentistry, Seoul 120-752, Korea
| | - Sien Lin
- Department of Orthopedic Surgery, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Yunzhi Peter Yang
- Department of Orthopedic Surgery, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA94305, USA
- Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, CA94305, USA
| |
Collapse
|
32
|
Łabowska MB, Cierluk K, Jankowska AM, Kulbacka J, Detyna J, Michalak I. A Review on the Adaption of Alginate-Gelatin Hydrogels for 3D Cultures and Bioprinting. MATERIALS (BASEL, SWITZERLAND) 2021; 14:858. [PMID: 33579053 PMCID: PMC7916803 DOI: 10.3390/ma14040858] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/12/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
Sustaining the vital functions of cells outside the organism requires strictly defined parameters. In order to ensure their optimal growth and development, it is necessary to provide a range of nutrients and regulators. Hydrogels are excellent materials for 3D in vitro cell cultures. Their ability to retain large amounts of liquid, as well as their biocompatibility, soft structures, and mechanical properties similar to these of living tissues, provide appropriate microenvironments that mimic extracellular matrix functions. The wide range of natural and synthetic polymeric materials, as well as the simplicity of their physico-chemical modification, allow the mechanical properties to be adjusted for different requirements. Sodium alginate-based hydrogel is a frequently used material for cell culture. The lack of cell-interactive properties makes this polysaccharide the most often applied in combination with other materials, including gelatin. The combination of both materials increases their biological activity and improves their material properties, making this combination a frequently used material in 3D printing technology. The use of hydrogels as inks in 3D printing allows the accurate manufacturing of scaffolds with complex shapes and geometries. The aim of this paper is to provide an overview of the materials used for 3D cell cultures, which are mainly alginate-gelatin hydrogels, including their properties and potential applications.
Collapse
Affiliation(s)
- Magdalena B. Łabowska
- Department of Mechanics, Materials and Biomedical Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland; (M.B.Ł); (A.M.J.)
| | - Karolina Cierluk
- Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373 Wroclaw, Poland;
| | - Agnieszka M. Jankowska
- Department of Mechanics, Materials and Biomedical Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland; (M.B.Ł); (A.M.J.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Jerzy Detyna
- Department of Mechanics, Materials and Biomedical Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland; (M.B.Ł); (A.M.J.)
| | - Izabela Michalak
- Department of Advanced Material Technologies, Faculty of Chemistry, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland;
| |
Collapse
|
33
|
Smith AM, Senior JJ. Alginate Hydrogels with Tuneable Properties. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 178:37-61. [PMID: 33547500 DOI: 10.1007/10_2020_161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Alginate is a material that has many biomedical applications due to its low toxicity and a variety of favourable physical properties. In particular, the ease in which hydrogels are formed from alginate and the variety of mechanical behaviours that can be imparted on the hydrogels, by understanding alginate chemistry and intuitive design, has made alginate the most widely investigated polysaccharide used for tissue engineering. This chapter provides an overview of alginate, from how the source and natural variations in composition can influence mechanical properties of alginate hydrogels, through to some innovative techniques used to modify and functionalise the hydrogels designed specifically for cell-based therapies. The main focus is on how these strategies of understanding and controlling the chemistry of alginates have resulted in the development of hydrogels that can be tuned to deliver the physical behaviours required for successful application. This will also highlight how research on the physicochemical properties has helped alginate evolve from a structural polysaccharide in brown seaweed into a highly tuneable, multifunctional, smart biomaterial, which is likely to find further biomedical applications in the future.
Collapse
Affiliation(s)
- Alan M Smith
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK.
| | - Jessica J Senior
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| |
Collapse
|
34
|
Mukherjee N, Adak A, Ghosh S. Recent trends in the development of peptide and protein-based hydrogel therapeutics for the healing of CNS injury. SOFT MATTER 2020; 16:10046-10064. [PMID: 32724981 DOI: 10.1039/d0sm00885k] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Traumatic brain injury (TBI) and spinal cord injury (SCI) cause millions of deaths and permanent or prolonged physical disabilities around the globe every year. It generally happens due to various incidents, such as accidents during sports, war, physical assault, and strokes which result in severe damage to brain and spinal cord. If this remains untreated, traumatic CNS injuries may lead to early development of several neurodegenerative diseases like Alzheimer's, Parkinson, multiple sclerosis, and other mental illnesses. The initial physical reaction, which is also termed as the primary phase, includes swelling, followed by inflammation as a result of internal haemorrhage causing damage to indigenous tissue, i.e., axonal shear injury, rupture of blood vessels, and partial impaired supply of oxygen and essential nutrients in the neurons, thereby initiating a cascade of events causing secondary injuries such as hypoxia, hypotension, cognitive impairment, seizures, imbalanced calcium homeostasis and glutamate-induced excitotoxicity resulting in concomitant neuronal cell death and cumulative permanent tissue damage. In the modern era of advanced biomedical technology, we are still living with scarcity of the clinically applicable comparative non-invasive therapeutic strategies for regeneration or functional recovery of neurons or neural networks after a massive CNS injury. One of the key reasons for this scarcity is the limited regenerative ability of neurons in CNS. Growth-impermissive glial scar and the lack of a synthetic biocompatible platform for proper neural tissue engineering and controlled supply of drugs further retard the healing process. Injectable or implantable hydrogel materials, consisting majorly of water in its porous three-dimensional (3D) structure, can serve as an excellent drug delivery platform as well as a transplanted cell-supporting scaffold medium. Among the various neuro-compatible bioinspired materials, we are limiting our discussion to the recent advancement of engineered biomaterials comprising mainly of peptides and proteins due to their growing demand, low immunogenicity and versatility in the fabrication of neuro regenerative medicine. In this article, we try to explore all the recent scientific avenues that are developing gradually to make peptide and peptide-conjugated biomaterial hydrogels as a therapeutic and supporting scaffold for treating CNS injuries.
Collapse
Affiliation(s)
- Nabanita Mukherjee
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India.
| | | | | |
Collapse
|
35
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
36
|
Belousov A, Titov S, Shved N, Malykin G, Kovalev V, Suprunova I, Khotimchenko Y, Kumeiko V. Hydrogels based on modified pectins capable of modulating neural cell behavior as prospective biomaterials in glioblastoma treatment. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:111-138. [PMID: 32448603 DOI: 10.1016/bs.irn.2020.03.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glioblastoma is the most common malignant tumor of the brain, but its treatment outcomes can be improved by new therapeutic techniques using biocompatible materials. Utilizing controllable alkaline de-esterification we obtained pectin preparation with 27.4% esterification degree and used it for bio-artificial matrix production. We discovered optimal gelation conditions in the presence of Ca2+ by the analysis of visco-elastic properties of the gels and produced a series of biomaterials in hydrogel forms. Hydrogels based on low-esterified pectin significantly slow down the metabolism of C6 glioma cells and neural stem cells (NSCs) and slightly decrease the viability of the C6 glioma, but not of NSCs. This happens due to a decrease in cell proliferation rate, while apoptosis degrees remain stable or negligibly decrease. We created a set of pectin hydrogels supplemented with different ratios of two ECM proteins-collagens I and IV. We have shown that the formation of cell processes in glioma C6 can be regulated by varying the ratio of two ECM proteins in gels used for 3D cell cultivation. Thus, composite matrix materials obtained can be used for modeling brain tumor invasion. The results presented suggest that modified pectins supplemented with two collagen types may serve as prospective biomaterials for glioblastoma treatment due to their ability to regulate glioma cell dynamics.
Collapse
Affiliation(s)
- Andrei Belousov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Sergei Titov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Nikita Shved
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Grigorii Malykin
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Valeri Kovalev
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Irina Suprunova
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Yuri Khotimchenko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia; Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia.
| |
Collapse
|
37
|
Fraczyk J, Wasko J, Walczak M, Kaminski ZJ, Puchowicz D, Kaminska I, Bogun M, Kolasa M, Stodolak-Zych E, Scislowska-Czarnecka A, Kolesinska B. Conjugates of Copper Alginate with Arginine-Glycine-Aspartic Acid (RGD) for Potential Use in Regenerative Medicine. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E337. [PMID: 31940765 PMCID: PMC7013949 DOI: 10.3390/ma13020337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 01/02/2023]
Abstract
Current restrictions on the use of antibiotics, associated with increases in bacterial resistance, require new solutions, including materials with antibacterial properties. In this study, copper alginate fibers obtained using the classic wet method were used to make nonwovens which were modified with arginine-glycine-aspartic acid (RGD) derivatives. Stable polysaccharide-peptide conjugates formed by coupling with 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium toluene-4-sulfonate (DMT/NMM/TosO-), and materials with physically embedded RGD derivatives, were obtained. The materials were found to be characterized by very high antibacterial activity against S. aureus and K. pneumoniae. Cytotoxicity studies confirmed that the materials are not cytotoxic. Copper alginate conjugates with RGD peptides have strong potential for use in regenerative medicine, due to their biocompatibility and innate antibacterial activity.
Collapse
Affiliation(s)
- Justyna Fraczyk
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90‐924 Lodz, Poland; (J.F.); (J.W.); (M.W.); (Z.J.K.)
| | - Joanna Wasko
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90‐924 Lodz, Poland; (J.F.); (J.W.); (M.W.); (Z.J.K.)
| | - Malgorzata Walczak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90‐924 Lodz, Poland; (J.F.); (J.W.); (M.W.); (Z.J.K.)
| | - Zbigniew J. Kaminski
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90‐924 Lodz, Poland; (J.F.); (J.W.); (M.W.); (Z.J.K.)
| | - Dorota Puchowicz
- Lukasiewicz Research Network-Textile Research Institute, Brzezinska 5/15, 92-103 Lodz, Poland; (D.P.); (I.K.); (M.B.)
| | - Irena Kaminska
- Lukasiewicz Research Network-Textile Research Institute, Brzezinska 5/15, 92-103 Lodz, Poland; (D.P.); (I.K.); (M.B.)
| | - Maciej Bogun
- Lukasiewicz Research Network-Textile Research Institute, Brzezinska 5/15, 92-103 Lodz, Poland; (D.P.); (I.K.); (M.B.)
| | - Marcin Kolasa
- Military Institute of Hygiene and Epidemiology Department of Pharmacology and Toxicology, Kozielska 4, 01-163 Warsaw, Poland;
| | - Ewa Stodolak-Zych
- Department of Biomaterials, AGH‐University of Science and Technology, A. Mickiewicz 30, 30-059 Krakow, Poland;
| | - Anna Scislowska-Czarnecka
- Academy of Physical Education, Department of Physiotherapy, Section of Anatomy, 31-008 Krakow, Poland;
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90‐924 Lodz, Poland; (J.F.); (J.W.); (M.W.); (Z.J.K.)
| |
Collapse
|
38
|
Growney EA, Linder HR, Garg K, Bledsoe JG, Sell SA. Bio-conjugation of platelet-rich plasma and alginate through carbodiimide chemistry for injectable hydrogel therapies. J Biomed Mater Res B Appl Biomater 2019; 108:1972-1984. [PMID: 31846217 DOI: 10.1002/jbm.b.34538] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/04/2019] [Accepted: 11/29/2019] [Indexed: 01/19/2023]
Abstract
Alginate is a highly tailorable, biocompatible polymer whose properties can be tuned to mimic the properties of native nucleus pulposus (NP) tissue. Platelet-rich plasma (PRP) is a highly accessible, inexpensive, and readily available mix of pro-regenerative factors. By functionalizing alginate with PRP, a mechanically optimized, bioactive alginate NP analogue may stimulate NP cells to proliferate and accumulate matrix over a longer period of time than if the PRP were solely encapsulated within the hydrogel. In this study, PRP was chemically bound to alginate using carbodiimide chemistry and mechanically, physically, and cytologically compared to plain alginate as well as alginate containing free-floating lyophilized PRP. The alginates were mechanically and physically characterized; PRP-conjugated alginate had similar mechanical properties to controls and had the benefit of retained PRP proteins within the hydrogel. Human nucleus pulposus cells (hNPCs) were seeded within the modified alginates and cultured for 14 days. Quantification data of glycosaminoglycans suggests that PRP-incorporated alginate has the potential to increase ECM production within the characterized alginate constructs, and that PRP-functionalized alginate can retain protein within the hydrogel over time. This is the first study to functionalize the milieu of PRP proteins onto alginate and characterize the mechanical and physical properties of the modified alginates. This study also incorporates hNPCs into the characterized PRP-modified alginates to observe phenotypic maintenance when encapsulated within the in situ gelling constructs.
Collapse
Affiliation(s)
- Emily A Growney
- Centre for Research in Medical Devices (CÙRAM), National University of Ireland Galway, Galway, Ireland.,Department of Biomedical Engineering, Parks College of Engineering, Aviation & Technology, Saint Louis University, St. Louis, Missouri
| | - Houston R Linder
- Department of Biomedical Engineering, Parks College of Engineering, Aviation & Technology, Saint Louis University, St. Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation & Technology, Saint Louis University, St. Louis, Missouri
| | - J Gary Bledsoe
- Department of Biomedical Engineering, Parks College of Engineering, Aviation & Technology, Saint Louis University, St. Louis, Missouri
| | - Scott A Sell
- Department of Biomedical Engineering, Parks College of Engineering, Aviation & Technology, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
39
|
Lewicki J, Bergman J, Kerins C, Hermanson O. Optimization of 3D bioprinting of human neuroblastoma cells using sodium alginate hydrogel. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.bprint.2019.e00053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
40
|
McCarthy RR, Ullah MW, Booth P, Pei E, Yang G. The use of bacterial polysaccharides in bioprinting. Biotechnol Adv 2019; 37:107448. [DOI: 10.1016/j.biotechadv.2019.107448] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
|
41
|
Patil S, Singh N. Silk fibroin-alginate based beads for human mesenchymal stem cell differentiation in 3D. Biomater Sci 2019; 7:4687-4697. [PMID: 31486468 DOI: 10.1039/c9bm01000a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lately silk fibroin has gained a lot of popularity as a tissue engineering scaffold due to its exceptional mechanical properties, negligible inflammatory reactions, remarkable biocompatibility, and tunable biodegradability. Nonetheless, 3 dimensional (3D) silk fibroin based scaffolds, which allow simultaneous formation of scaffolds and cell encapsulation with minimal damage to the cells, are unavailable, as most of the methods involve the use of some cell destructive techniques. Thus, cells have to be loaded after the scaffold formation and the study has to rely upon the ability of the cells to penetrate the scaffold to obtain a 3D microenvironment. Hence, these platforms do not allow for a true 3D system replicating the in vivo environment. Here silk fibroin-alginate based beads have been developed, and retain silk fibroin for a longer period of time and allow for simultaneous cell encapsulation as the crosslinking method is cell-compatible. It is demonstrated for the first time that these silk fibroin-alginate beads can be used to encapsulate the cells at varying cell densities depending on the desired application. These beads were further used to study the effect of functional groups on human mesenchymal stem cell (hMSC) differentiation in 3D, by utilizing carboxylic groups naturally present in alginate as well as introducing phosphate groups. The results showed that these beads were able to support the growth and proliferation of hMSCs and induced differentiation solely due to functional groups within 14 days. These beads were better in directing hMSC differentiation into osteogenic and chondrogenic lineages compared to 2D surfaces and differentiation media.
Collapse
Affiliation(s)
- Smita Patil
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India. and Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
42
|
Yao R, Alkhawtani AYF, Chen R, Luan J, Xu M. Rapid and efficient in vivo angiogenesis directed by electro-assisted bioprinting of alginate/collagen microspheres with human umbilical vein endothelial cell coating layer. Int J Bioprint 2019; 5:194. [PMID: 32596542 PMCID: PMC7310271 DOI: 10.18063/ijb.v5i2.1.194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
Rapid reconstruction of functional microvasculature is the urgent challenge of regenerative medicine and ischemia therapy development. The purpose of this study was to provide an alternative solution for obtaining functional blood vessel networks in vivo, through assessing whether hydrogel-based microspheres coated by human umbilical vein endothelial cells (HUVECs) can direct rapid and efficient in vivo angiogenesis without the addition of exogenous growth factors or other supporting cells. Uniform alginate microspheres with adjustable diameter were biofabricated by electro-assisted bioprinting technology. Collagen fibrils were evenly coated on the surface of alginate microspheres through simple self-assembly procedure, and collagen concentration is optimized to achieve the highest HUVECs adhesion and proliferation. Immunofluorescence staining and gene analysis confirmed the formation of the prevascularized tubular structure and significantly enhanced endothelial gene expression. HUVECs-coated hydrogel microspheres with different diameters were subcutaneously injected in immune-deficient mice, which demonstrated rapid blood vessel regeneration and functional anastomosis with host blood vessels within 1 week. Besides, microsphere diameter demonstrated influence on blood vessel density with statistical differences but showed no obvious influence on the area occupied by blood vessels. This study provided a powerful tool for rapid and minimal-invasion angiogenesis of bioprinting constructs and a potential method for vascularized tissue regeneration and ischemia treatment with clinically relevant dimensions.
Collapse
Affiliation(s)
- Rui Yao
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People’s Republic of China
| | - Ahmed Yousef F. Alkhawtani
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People’s Republic of China
| | - Ruoyu Chen
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, People’s Republic of China
| | - Jie Luan
- Plastic Surgery Hospital, Peking Union Medical College, Beijing, 100144, People’s Republic of China
| | - Mingen Xu
- Key Laboratory of Medical Information and Three-dimensional Bioprinting of Zhejiang Province, Hangzhou Dianzi University, Hangzhou 310018, China
| |
Collapse
|
43
|
Bio-fabrication of peptide-modified alginate scaffolds: Printability, mechanical stability and neurite outgrowth assessments. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.bprint.2019.e00045] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
44
|
Seidlits SK, Liang J, Bierman RD, Sohrabi A, Karam J, Holley SM, Cepeda C, Walthers CM. Peptide-modified, hyaluronic acid-based hydrogels as a 3D culture platform for neural stem/progenitor cell engineering. J Biomed Mater Res A 2019; 107:704-718. [PMID: 30615255 PMCID: PMC8862560 DOI: 10.1002/jbm.a.36603] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/23/2018] [Accepted: 01/03/2019] [Indexed: 07/26/2023]
Abstract
Neural stem/progenitor cell (NS/PC)-based therapies have shown exciting potential for regeneration of the central nervous system (CNS) and NS/PC cultures represent an important resource for disease modeling and drug screening. However, significant challenges limiting clinical translation remain, such as generating large numbers of cells required for model cultures or transplantation, maintaining physiologically representative phenotypes ex vivo and directing NS/PC differentiation into specific fates. Here, we report that culture of human NS/PCs in 3D, hyaluronic acid (HA)-rich biomaterial microenvironments increased differentiation toward oligodendrocytes and neurons over 2D cultures on laminin-coated glass. Moreover, NS/PCs in 3D culture exhibited a significant reduction in differentiation into reactive astrocytes. Many NS/PC-derived neurons in 3D, HA-based hydrogels expressed synaptophysin, indicating synapse formation, and displayed electrophysiological characteristics of immature neurons. While inclusion of integrin-binding, RGD peptides into hydrogels resulted in a modest increase in numbers of viable NS/PCs, no combination of laminin-derived, adhesive peptides affected differentiation outcomes. Notably, 3D cultures of differentiating NS/PCs were maintained for at least 70 days in medium with minimal growth factor supplementation. In sum, results demonstrate the use of 3D, HA-based biomaterials for long-term expansion and differentiation of NS/PCs toward oligodendroglial and neuronal fates, while inhibiting astroglial fates. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 704-718, 2019.
Collapse
Affiliation(s)
- Stephanie K. Seidlits
- Department of Bioengineering, UCLA, Los Angels, California
- Board Stem Cell Research Center, UCLA, Los Angels, California
- Brain Research Institute, UCLA, Los Angels, California
- Jonsson Comprehensive Cancer Center, UCLA, Los Angels, California
- Center for Minimally Invasive Therapeutics, UCLA, Los Angels, California
| | - Jesse Liang
- Department of Bioengineering, UCLA, Los Angels, California
| | | | | | - Joshua Karam
- Department of Bioengineering, UCLA, Los Angels, California
| | - Sandra M. Holley
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | | |
Collapse
|
45
|
George J, Hsu CC, Nguyen LTB, Ye H, Cui Z. Neural tissue engineering with structured hydrogels in CNS models and therapies. Biotechnol Adv 2019; 42:107370. [PMID: 30902729 DOI: 10.1016/j.biotechadv.2019.03.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/25/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
Abstract
The development of techniques to create and use multiphase microstructured hydrogels (granular hydrogels or microgels) has enabled the generation of cultures with more biologically relevant architecture and use of structured hydrogels is especially pertinent to the development of new types of central nervous system (CNS) culture models and therapies. We review material choice and the customisation of hydrogel structure, as well as the use of hydrogels in developmental models. Combining the use of structured hydrogel techniques with developmentally relevant tissue culture approaches will enable the generation of more relevant models and treatments to repair damaged CNS tissue architecture.
Collapse
Affiliation(s)
- Julian George
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Chia-Chen Hsu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Linh Thuy Ba Nguyen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
46
|
Severino P, da Silva CF, Andrade LN, de Lima Oliveira D, Campos J, Souto EB. Alginate Nanoparticles for Drug Delivery and Targeting. Curr Pharm Des 2019; 25:1312-1334. [PMID: 31465282 DOI: 10.2174/1381612825666190425163424] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/15/2019] [Indexed: 12/31/2022]
Abstract
Nanotechnology refers to the control, manipulation, study and manufacture of structures and devices at the nanometer size range. The small size, customized surface, improved solubility and multi-functionality of nanoparticles will continue to create new biomedical applications, as nanoparticles allow to dominate stability, solubility and bioavailability, as well controlled release of drugs. The type of a nanoparticle, and its related chemical, physical and morphological properties influence its interaction with living cells, as well as determine the route of clearance and possible toxic effects. This field requires cross-disciplinary research and gives opportunities to design and develop multifunctional devices, which allow the diagnosis and treatment of devastating diseases. Over the past few decades, biodegradable polymers have been studied for the fabrication of drug delivery systems. There was extensive development of biodegradable polymeric nanoparticles for drug delivery and tissue engineering, in view of their applications in controlling the release of drugs, stabilizing labile molecules from degradation and site-specific drug targeting. The primary aim is to reduce dosing frequency and prolong the therapeutic outcomes. For this purpose, inert excipients should be selected, being biopolymers, e.g. sodium alginate, commonly used in controlled drug delivery. Nanoparticles composed of alginate (known as anionic polysaccharide widely distributed in the cell walls of brown algae which, when in contact with water, forms a viscous gum) have emerged as one of the most extensively characterized biomaterials used for drug delivery and targeting a set of administration routes. Their advantages include not only the versatile physicochemical properties, which allow chemical modifications for site-specific targeting but also their biocompatibility and biodegradation profiles, as well as mucoadhesiveness. Furthermore, mechanical strength, gelation, and cell affinity can be modulated by combining alginate nanoparticles with other polymers, surface tailoring using specific targeting moieties and by chemical or physical cross-linking. However, for every physicochemical modification in the macromolecule/ nanoparticles, a new toxicological profile may be obtained. In this paper, the different aspects related to the use of alginate nanoparticles for drug delivery and targeting have been revised, as well as how their toxicological profile will determine the therapeutic outcome of the drug delivery system.
Collapse
Affiliation(s)
- Patricia Severino
- Universidade Tiradentes (Unit), Av. Murilo Dantas, 300, Farolandia, Aracaju-SE, CEP 49.032-490, Brazil
- Instituto de Tecnologia e Pesquisa, Laboratório de Nanotecnologia e Nanomedicina (LNMed) Av. Murilo Dantas, 300, Aracaju - SE, CEP 49.032-490, Brazil
| | - Classius F da Silva
- Universidade Federal de Sao Paulo, Instituto de Ciências Ambientais, Quimicas e Farmaceuticas, Departamento de Engenharia Quimica, Rua Sao Nicolau, 210, Diadema - SP, CEP 09.913-030, Brazil
| | - Luciana N Andrade
- Universidade Tiradentes (Unit), Av. Murilo Dantas, 300, Farolandia, Aracaju-SE, CEP 49.032-490, Brazil
- Instituto de Tecnologia e Pesquisa, Laboratório de Nanotecnologia e Nanomedicina (LNMed) Av. Murilo Dantas, 300, Aracaju - SE, CEP 49.032-490, Brazil
| | - Daniele de Lima Oliveira
- Universidade Tiradentes (Unit), Av. Murilo Dantas, 300, Farolandia, Aracaju-SE, CEP 49.032-490, Brazil
- Instituto de Tecnologia e Pesquisa, Laboratório de Nanotecnologia e Nanomedicina (LNMed) Av. Murilo Dantas, 300, Aracaju - SE, CEP 49.032-490, Brazil
| | - Joana Campos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC), Polo das Ciencias da Saude, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC), Polo das Ciencias da Saude, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
| |
Collapse
|
47
|
Polycaprolactone porous template facilitates modulated release of molecules from alginate hydrogels. REACT FUNCT POLYM 2018. [DOI: 10.1016/j.reactfunctpolym.2018.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
48
|
Schulz A, Gepp MM, Stracke F, von Briesen H, Neubauer JC, Zimmermann H. Tyramine-conjugated alginate hydrogels as a platform for bioactive scaffolds. J Biomed Mater Res A 2018; 107:114-121. [PMID: 30256518 PMCID: PMC6585978 DOI: 10.1002/jbm.a.36538] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/14/2018] [Accepted: 08/29/2018] [Indexed: 02/04/2023]
Abstract
Alginate‐based hydrogels represent promising microenvironments for cell culture and tissue engineering, as their mechanical and porous characteristics are adjustable toward in vivo conditions. However, alginate scaffolds are bioinert and thus inhibit cellular interactions. To overcome this disadvantage, bioactive alginate surfaces were produced by conjugating tyramine molecules to high‐molecular‐weight alginates using the carbodiimide chemistry. Structural elucidation using nuclear magnetic resonance spectroscopy and contact angle measurements revealed a surface chemistry and wettability of tyramine‐alginate hydrogels similar to standard cell culture treated polystyrene. In contrast to stiff cell culture plastic, tyramine‐alginate scaffolds were found to be soft (60–80 kPa), meeting the elastic moduli of human tissues such as liver and heart. We further demonstrated an enhanced protein adsorption with increasing tyramine conjugation, stable for several weeks. Cell culture studies with human mesenchymal stem cells and human pluripotent stem cell‐derived cardiomyocytes qualified tyramine‐alginate hydrogels as bioactive platforms enabling cell adhesion and contraction on (structured) 2‐D layer and spherical matrices. Due to the alginate functionalization with tyramines, stable cell–matrix interactions were observed beneficial for an implementation in biology, biotechnology, and medicine toward efficient cell culture and tissue substitutes. © 2018 The Authors. Journal of Biomedical Materials Research Part A published by Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 114–121, 2019.
Collapse
Affiliation(s)
- André Schulz
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, 66280, Germany
| | - Michael M Gepp
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, 66280, Germany.,Fraunhofer Project Center for Stem Cell Process Engineering, Wuerzburg, 97082, Germany
| | - Frank Stracke
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, 66280, Germany
| | - Hagen von Briesen
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, 66280, Germany
| | - Julia C Neubauer
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, 66280, Germany.,Fraunhofer Project Center for Stem Cell Process Engineering, Wuerzburg, 97082, Germany
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, 66280, Germany.,Chair for Molecular and Cellular Biotechnology, Saarland University, Saarbruecken, 66123, Germany.,Faculty of Marine Science, Universidad Católica del Norte, Coquimbo, Chile
| |
Collapse
|
49
|
Ochbaum G, Davidovich-Pinhas M, Bitton R. Tuning the mechanical properties of alginate-peptide hydrogels. SOFT MATTER 2018; 14:4364-4373. [PMID: 29781028 DOI: 10.1039/c8sm00059j] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Alginate, a polysaccharide that gels in the presence of divalent ions, has been used in the field of regenerative medicine to facilitate cell growth in impaired tissues by providing an artificial bio-surrounding similar to the natural extra cellular matrix (ECM). Here, we present a systematic investigation of the effect of three arginine-glycine-aspartic acid (RGD)-containing peptides, G6KRGDY, A6KRGDY and V6KRGDY, on the physical properties of alginate-peptide hydrogels. Rheology measurements showed that the storage modulus of the alginate-A6KRGDY and alginate-V6KRGDY gels is an order of magnitude higher than that of the alginate-G6KRGDY gel. Small angle X-ray scattering (SAXS) measurements suggest that the difference in the mechanical properties of the gels is due to the formation of larger peptide junction zones in addition to the ones formed by calcium ions. These findings indicate that the peptides' ability to self-assemble in aqueous solution is a significant factor in tuning the stiffness of the alginate/peptide hybrid hydrogels.
Collapse
Affiliation(s)
- Guy Ochbaum
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | | | | |
Collapse
|
50
|
Joo S, Song SY, Nam YS, Nam Y. Stimuli-Responsive Neuronal Networking via Removable Alginate Masks. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Sunghoon Joo
- Department of Bio and Brain Engineering; Korea Advanced Institute of Science and Technology (KAIST); Daejeon 34141 Republic of Korea
| | - Seuk Young Song
- Department of Materials Science and Engineering; Korea Advanced Institute of Science and Technology (KAIST); Daejeon 34141 Republic of Korea
| | - Yoon Sung Nam
- Department of Materials Science and Engineering; Korea Advanced Institute of Science and Technology (KAIST); Daejeon 34141 Republic of Korea
- KAIST Institute for the NanoCentury; Korea Advanced Institute of Science and Technology (KAIST); Daejeon 34141 Republic of Korea
| | - Yoonkey Nam
- Department of Bio and Brain Engineering; Korea Advanced Institute of Science and Technology (KAIST); Daejeon 34141 Republic of Korea
- KAIST Institute for the NanoCentury; Korea Advanced Institute of Science and Technology (KAIST); Daejeon 34141 Republic of Korea
| |
Collapse
|