1
|
Miller RC, Temenoff JS. Biomaterials for Cell Manufacturing. ACS Macro Lett 2024; 13:1521-1530. [PMID: 39466845 PMCID: PMC11580378 DOI: 10.1021/acsmacrolett.4c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Cell therapies, potent populations of cells used to treat disease and injury, can be strategically manufactured with biomaterial intervention to improve clinical translation. In this viewpoint, we discuss biomaterial design and integration into cell manufacturing steps to achieve three main goals: scale-up, phenotype control, and selection of potent cells. Material properties can be engineered to influence the cell-biomaterial interface and, therefore, impart desirable cell behavior such as growth, secretory activity, and differentiation. Future directions for the field should capitalize on the combinatorial design of biomaterial properties to yield highly specific and potent cell populations. Furthermore, future biomaterials could contribute to novel high-throughput cell separation technologies that can individually select the most therapeutically relevant cells within a produced batch.
Collapse
Affiliation(s)
- Ryan C. Miller
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech/Emory University, Atlanta, Georgia 30332, United States
| | - Johnna S. Temenoff
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech/Emory University, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
2
|
Baines DK, Platania V, Tavernaraki NN, Parati M, Wright K, Radecka I, Chatzinikolaidou M, Douglas TEL. The Enrichment of Whey Protein Isolate Hydrogels with Poly-γ-Glutamic Acid Promotes the Proliferation and Osteogenic Differentiation of Preosteoblasts. Gels 2023; 10:18. [PMID: 38247741 PMCID: PMC10815088 DOI: 10.3390/gels10010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Osseous disease accounts for over half of chronic pathologies, but there is a limited supply of autografts, the gold standard; hence, there is a demand for new synthetic biomaterials. Herein, we present the use of a promising, new dairy-derived biomaterial: whey protein isolate (WPI) in the form of hydrogels, modified with the addition of different concentrations of the biotechnologically produced protein-like polymeric substance poly-γ-glutamic acid (γ-PGA) as a potential scaffold for tissue regeneration. Raman spectroscopic analysis demonstrated the successful creation of WPI-γ-PGA hydrogels. A cytotoxicity assessment using preosteoblastic cells demonstrated that the hydrogels were noncytotoxic and supported cell proliferation from day 3 to 14. All γ-PGA-containing scaffold compositions strongly promoted cell attachment and the formation of dense interconnected cell layers. Cell viability was significantly increased on γ-PGA-containing scaffolds on day 14 compared to WPI control scaffolds. Significantly, the cells showed markers of osteogenic differentiation; they synthesised increasing amounts of collagen over time, and cells showed significantly enhanced alkaline phosphatase activity at day 7 and higher levels of calcium for matrix mineralization at days 14 and 21 on the γ-PGA-containing scaffolds. These results demonstrated the potential of WPI-γ-PGA hydrogels as scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Daniel K. Baines
- Faculty of Science and Technology, School of Engineering, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
- Faculty of Health and medicine, Division of Biomedical and Life Sciences, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| | - Varvara Platania
- Department of Materials Science and Technology, University of Crete, GR-70013 Heraklion, Greece; (V.P.); (N.N.T.); (M.C.)
| | - Nikoleta N. Tavernaraki
- Department of Materials Science and Technology, University of Crete, GR-70013 Heraklion, Greece; (V.P.); (N.N.T.); (M.C.)
| | - Mattia Parati
- Faculty of Science and Engineering, School of Life Sciences, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.P.); (I.R.)
| | - Karen Wright
- Faculty of Health and medicine, Division of Biomedical and Life Sciences, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| | - Iza Radecka
- Faculty of Science and Engineering, School of Life Sciences, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.P.); (I.R.)
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, GR-70013 Heraklion, Greece; (V.P.); (N.N.T.); (M.C.)
- Institute of Electronic Structure and Laser, Foundation for Research and Technology Hellas, GR-70013 Heraklion, Greece
| | - Timothy E. L. Douglas
- Faculty of Science and Technology, School of Engineering, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| |
Collapse
|
3
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [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: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
4
|
Tarricone G, Carmagnola I, Chiono V. Tissue-Engineered Models of the Human Brain: State-of-the-Art Analysis and Challenges. J Funct Biomater 2022; 13:146. [PMID: 36135581 PMCID: PMC9501967 DOI: 10.3390/jfb13030146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Neurological disorders affect billions of people across the world, making the discovery of effective treatments an important challenge. The evaluation of drug efficacy is further complicated because of the lack of in vitro models able to reproduce the complexity of the human brain structure and functions. Some limitations of 2D preclinical models of the human brain have been overcome by the use of 3D cultures such as cell spheroids, organoids and organs-on-chip. However, one of the most promising approaches for mimicking not only cell structure, but also brain architecture, is currently represented by tissue-engineered brain models. Both conventional (particularly electrospinning and salt leaching) and unconventional (particularly bioprinting) techniques have been exploited, making use of natural polymers or combinations between natural and synthetic polymers. Moreover, the use of induced pluripotent stem cells (iPSCs) has allowed the co-culture of different human brain cells (neurons, astrocytes, oligodendrocytes, microglia), helping towards approaching the central nervous system complexity. In this review article, we explain the importance of in vitro brain modeling, and present the main in vitro brain models developed to date, with a special focus on the most recent advancements in tissue-engineered brain models making use of iPSCs. Finally, we critically discuss achievements, main challenges and future perspectives.
Collapse
Affiliation(s)
- Giulia Tarricone
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
- Department of Chemistry and Industrial Chemistry, University of Genova, Via Dodecaneso 31, 16146 Genova, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| |
Collapse
|
5
|
Cho SH, Shin KK, Kim SY, Cho MY, Oh DB, Lim YT. In Situ-Forming Collagen/poly-γ-glutamic Acid Hydrogel System with Mesenchymal Stem Cells and Bone Morphogenetic Protein-2 for Bone Tissue Regeneration in a Mouse Calvarial Bone Defect Model. Tissue Eng Regen Med 2022; 19:1099-1111. [PMID: 35460494 PMCID: PMC9477999 DOI: 10.1007/s13770-022-00454-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/21/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Bone marrow-derived mesenchymal stem cells (BMSCs) and bone morphogenetic protein-2 (BMP-2) have been studied for bone repair because they have regenerative potential to differentiate into osteoblasts. The development of injectable and in situ three-dimensional (3D) scaffolds to proliferate and differentiate BMSCs and deliver BMP-2 is a crucial technology in BMSC-based tissue engineering. METHODS The proliferation of mouse BMSCs (mBMSCs) in collagen/poly-γ-glutamic acid (Col/γ-PGA) hydrogel was evaluated using LIVE/DEAD and acridine orange and propidium iodide assays. In vitro osteogenic differentiation and the gene expression level of Col/γ-PGA(mBMSC/BMP-2) were assessed by alizarin red S staining and quantitative reverse-transcription polymerase chain reaction. The bone regeneration effect of Col/γ-PGA(mBMSC/BMP-2) was evaluated in a mouse calvarial bone defect model. The cranial bones of the mice were monitored by micro-computed tomography and histological analysis. RESULTS The developed Col/γ-PGA hydrogel showed low viscosity below ambient temperature, while it provided a high elastic modulus and viscous modulus at body temperature. After gelation, the Col/γ-PGA hydrogel showed a 3D and interconnected porous structure, which helped the effective proliferation of BMSCs with BMP-2. The Col/γ-PGA (mBMSC/BMP-2) expressed more osteogenic genes and showed effective orthotopic bone formation in a mouse model with a critical-sized bone defect in only 3-4 weeks. CONCLUSION The Col/γ-PGA(mBMSC/BMP-2) hydrogel was suggested to be a promising platform by combining collagen as a major component of the extracellular matrix and γ-PGA as a viscosity reducer for easy handling at room temperature in BMSC-based bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Sun-Hee Cho
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Chungcheongbuk-do, 28119, Republic of Korea
| | - Keun Koo Shin
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Sun-Young Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Mi Young Cho
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Chungcheongbuk-do, 28119, Republic of Korea
| | - Doo-Byoung Oh
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.
- Department of Nano Engineering and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
6
|
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
|
7
|
Park SB, Sung MH, Uyama H, Han DK. Poly(glutamic acid): Production, composites, and medical applications of the next-generation biopolymer. Prog Polym Sci 2021. [DOI: 10.1016/j.progpolymsci.2020.101341] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
8
|
Vila-Parrondo C, García-Astrain C, Liz-Marzán LM. Colloidal systems toward 3D cell culture scaffolds. Adv Colloid Interface Sci 2020; 283:102237. [PMID: 32823220 DOI: 10.1016/j.cis.2020.102237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
Three-dimensional porous scaffolds are essential for the development of tissue engineering and regeneration, as biomimetic supports to recreate the microenvironment present in natural tissues. To successfully achieve the growth and development of a specific kind of tissue, porous matrices should be able to influence cell behavior by promoting close cell-cell and cell-matrix interactions. To achieve this goal, the scaffold must fulfil a set of conditions, including ordered interconnected porosity to promote cell diffusion and vascularization, mechanical strength to support the tissue during continuous ingrowth, and biocompatibility to avoid toxicity. Among various building approaches to the construction of porous matrices, selected strategies afford hierarchical scaffolds with such defined properties. The control over porosity, microstructure or morphology, is crucial to the fabrication of high-end, reproducible scaffolds for the target application. In this review, we provide an insight into recent advances toward the colloidal fabrication of hierarchical scaffolds. After identifying the main requirements for scaffolds in biomedical applications, conceptual building processes are introduced. Examples of tissue regeneration applications are provided for different scaffold types, highlighting their versatility and biocompatibility. We finally provide a prospect about the current state of the art and limitations of porous scaffolds, along with challenges that are to be addressed, so these materials consolidate in the fields of tissue engineering and drug delivery.
Collapse
|
9
|
Revkova VA, Grebenik EA, Kalsin VA, Demina TS, Bardakova KN, Shavkuta BS, Melnikov PA, Samoilova EM, Konoplyannikov MA, Efremov YM, Zhang C, Akopova TA, Troitsky AV, Timashev PS, Baklaushev VP. Chitosan- g-oligo(L,L-lactide) Copolymer Hydrogel Potential for Neural Stem Cell Differentiation. Tissue Eng Part A 2020; 26:953-963. [PMID: 32159465 DOI: 10.1089/ten.tea.2019.0265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We evaluated the applicability of chitosan-g-oligo(L,L-lactide) copolymer (CLC) hydrogel for central nervous system tissue engineering. The biomechanical properties of the CLC hydrogel were characterized and its biocompatibility was assessed with neural progenitor cells obtained from two different sources: H9-derived neural stem cells (H9D-NSCs) and directly reprogrammed neural precursor cells (drNPCs). Our study found that the optically transparent CLC hydrogel possessed biomechanical characteristics suitable for culturing human neural stem/precursor cells and was noncytotoxic. When seeded on films prepared from CLC copolymer hydrogel, both H9D-NSC and drNPC adhered well, expanded and exhibited signs of spontaneous differentiation. While H9D-NSC mainly preserved multipotency as shown by a high proportion of Nestin+ and Sox2+ cells and a comparatively lower expression of the neuronal markers βIII-tubulin and MAP2, drNPCs, obtained by direct reprogramming, differentiated more extensively along the neuronal lineage. Our study indicates that the CLC hydrogel may be considered as a substrate for tissue-engineered constructs, applicable for therapy of neurodegenerative diseases. Impact statement We synthetized a chitosan-g-oligo(L,L-lactide) hydrogel that sustained multipotency of embryonic-derived neural stem cells (NSCs) and supported differentiation of directly reprogrammed NSC predominantly along the neuronal lineage. The hydrogel exhibited no cytotoxicity in vitro, both in extraction and contact cytotoxicity tests. When seeded on the hydrogel, both types of NSCs adhered well, expanded, and exhibited signs of spontaneous differentiation. The biomechanical properties of the hydrogel were similar to that of human spinal cord with incised pia mater. These data pave the way for further investigations of the hydrogel toward its applicability in central nervous system tissue engineering.
Collapse
Affiliation(s)
- Veronica A Revkova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Ekaterina A Grebenik
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir A Kalsin
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Tatiana S Demina
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Enikolopov Institute of Synthetic Polymer Materials, Russian Academy of Sciences, Moscow, Russia
| | - Kseniia N Bardakova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Research Center "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Boris S Shavkuta
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Research Center "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Pavel A Melnikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia.,Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Ekaterina M Samoilova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Mikhail A Konoplyannikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yuri M Efremov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumors, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tatiana A Akopova
- Enikolopov Institute of Synthetic Polymer Materials, Russian Academy of Sciences, Moscow, Russia
| | - Alexandr V Troitsky
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Research Center "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia.,N.N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir P Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| |
Collapse
|
10
|
Velmurugan BK, Bharathi Priya L, Poornima P, Lee LJ, Baskaran R. Biomaterial aided differentiation and maturation of induced pluripotent stem cells. J Cell Physiol 2018; 234:8443-8454. [PMID: 30565686 DOI: 10.1002/jcp.27769] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022]
Abstract
Engineering/reprogramming differentiated adult somatic cells to gain the ability to differentiate into any type of cell lineage are called as induced pluripotent stem cells (iPSCs). Offering unlimited self-renewal and differentiation potential, these iPSC are aspired to meet the growing demands in the field of regenerative medicine, tissue engineering, disease modeling, nanotechnology, and drug discovery. Biomaterial fabrication with the rapid evolution of technology increased their versatility and utility in regenerative medicine and tissue engineering, revolutionizing the stem cell biology research with the property to guide the process of proliferation, differentiation, and morphogenesis. Combining traditional culture platforms of iPSC with biomaterials aids to overcome the limitations associated with derivation, proliferation, and maturation, thereby could improve the clinical translation of iPSC. The present review discusses in brief about the reprogramming techniques for the derivation iPSC and details on several biomaterial guided differentiation of iPSC to different cell types with specific relevance to tissue engineering/regenerative medicine.
Collapse
Affiliation(s)
| | - Lohanathan Bharathi Priya
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Paramasivan Poornima
- Molecular and Cellular Pharmacology Laboratory, School of Science, Engineering and Technology, University of Abertay, Dundee, UK
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rathinasamy Baskaran
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
11
|
Kornev VA, Grebenik EA, Solovieva AB, Dmitriev RI, Timashev PS. Hydrogel-assisted neuroregeneration approaches towards brain injury therapy: A state-of-the-art review. Comput Struct Biotechnol J 2018; 16:488-502. [PMID: 30455858 PMCID: PMC6232648 DOI: 10.1016/j.csbj.2018.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022] Open
Abstract
Recent years have witnessed the development of an enormous variety of hydrogel-based systems for neuroregeneration. Formed from hydrophilic polymers and comprised of up to 90% of water, these three-dimensional networks are promising tools for brain tissue regeneration. They can assist structural and functional restoration of damaged tissues by providing mechanical support and navigating cell fate. Hydrogels also show the potential for brain injury therapy due to their broadly tunable physical, chemical, and biological properties. Hydrogel polymers, which have been extensively implemented in recent brain injury repair studies, include hyaluronic acid, collagen type I, alginate, chitosan, methylcellulose, Matrigel, fibrin, gellan gum, self-assembling peptides and proteins, poly(ethylene glycol), methacrylates, and methacrylamides. When viewed as tools for neuroregeneration, hydrogels can be divided into: (1) hydrogels suitable for brain injury therapy, (2) hydrogels that do not meet basic therapeutic requirements and (3) promising hydrogels which meet the criteria for further investigations. Our analysis shows that fibrin, collagen I and self-assembling peptide-based hydrogels display very attractive properties for neuroregeneration.
Collapse
Affiliation(s)
- Vladimir A. Kornev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Ekaterina A. Grebenik
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Anna B. Solovieva
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
| | - Ruslan I. Dmitriev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
- Institute of Photonic Technologies, Research Center “Crystallography and Photonics” Russian Academy of Sciences, 2 Pionerskaya st., Troitsk, Moscow 108840, Russian Federation
| |
Collapse
|
12
|
Luo D, Ruan S, Liu A, Kong X, Lee IS, Chen C. Laminin functionalized biomimetic apatite to regulate the adhesion and proliferation behaviors of neural stem cells. Int J Nanomedicine 2018; 13:6223-6233. [PMID: 30349246 PMCID: PMC6188167 DOI: 10.2147/ijn.s176596] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Functionalizing biomaterial substrates with biological signals shows promise in regulating neural stem cell (NSC) behaviors through mimicking cellular microenvironment. However, diverse methods for immobilizing biological molecules yields promising results but with many problems. Biomimetic apatite is an excellent carrier due to its non-toxicity, good biocompatibility, biodegradability, and favorable affinity to plenty of molecules. Therefore, it may provide a promising alternative in regulating NSC behaviors. Methods Biomimetic apatite immobilized with the extracellular protein - laminin (LN) was prepared through coprecipitation process in modified Dulbecco's phosphate-buffered saline (DPBS) containing LN. The amount of coprecipitated LN and their release kinetics were examined. The adhesion and proliferation behaviors of NSC on biomimetic apatite immobilized with LN were investigated. Results The coprecipitation approach provided well retention of LN within biomimetic apatite up to 28 days, and supported the adhesion and proliferation of NSCs without cytotoxicity. For long-term cultivation, NSCs formed neurosphere-like aggregates on non-functionalized biomimetic apatite. A monolayer of proliferated NSCs on biomimetic apatite with coprecipitated LN was observed and even more stable than the positive control of LN coated tissue-culture treated polystyrene (TCP). Conclusion The simple and reproducible method of coprecipitation suggests that biomimetic apatite is an ideal carrier to functionalize materials with biological molecules for neural-related applications.
Collapse
Affiliation(s)
- Dandan Luo
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China,
| | - Shichao Ruan
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China,
| | - Aiping Liu
- Center for Optoelectronics Materials and Devices, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Xiangdong Kong
- College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China,
| | - In-Seop Lee
- College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China, .,Institute of Natural Sciences, Yonsei University, Seoul 03722, Korea,
| | - Cen Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China, .,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, People's Republic of China,
| |
Collapse
|
13
|
Hydrogel Scaffolds: Towards Restitution of Ischemic Stroke-Injured Brain. Transl Stroke Res 2018; 10:1-18. [DOI: 10.1007/s12975-018-0655-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/17/2018] [Accepted: 08/19/2018] [Indexed: 12/27/2022]
|
14
|
Chen YS, Harn HJ, Chiou TW. The Role of Biomaterials in Implantation for Central Nervous System Injury. Cell Transplant 2018; 27:407-422. [PMID: 29741115 PMCID: PMC6038039 DOI: 10.1177/0963689717732991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Permanent deficits that occur in memory, sensation, and cognition can result from central nervous system (CNS) trauma that causes dysfunction and/or unregulated CNS regeneration. Some therapeutic approaches are preferentially applied to the human body. Therefore, cell transplantation, one of the therapeutic strategies, may be used to benefit people. However, poor cell viability and low efficacy are the limitations to cell transplantation strategies. Biomaterials have been widely used in several fields (e.g., triggering cell differentiation, guiding cell migration, improving wound healing, and increasing tissue regeneration) by modulating their characteristics in chemistry, topography, and softness/stiffness for highly flexible application. We reviewed implanted biomaterials to investigate the roles and influences of physical/chemical properties on cell behaviors and applications. With their unique molecular features, biomaterials are delivered in several methods and mixed with transplanted cells, which assists in increasing postimplanted biological substance efficiency on cell survival, host responses, and functional recovery of animal models. Moreover, tracking the routes of these transplanted cells using biomaterials as labeling agents is crucial for addressing their location, distribution, activity, and viability. Here, we provide comprehensive comments and up-to-date research of the application of biomaterials.
Collapse
Affiliation(s)
- Yu-Shuan Chen
- Bioinnovation Center, Tzu Chi Foundation, Hualien, Taiwan, No. 707, Sec. 3, Chung Yang Rd., Hualien 970, Taiwan, R.O.C.,Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, No. 707, Section 3, Chung-Yang Road, Hualien 970, Taiwan, R.O.C
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi foundation, Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, 707, Sec. 3, Chung Yang Rd., Hualien 970, Taiwan, R.O.C.,Horng-Jyh Harn, MD, PhD, Bioinnovation Center, Tzu Chi foundation, Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, 707, Sec. 3, Chung Yang Rd., Hualien 970, Taiwan, R.O.C.
| | - Tzyy-Wen Chiou
- Department of Life Science, Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien 97401, Taiwan, R.O.C.,Tzyy-Wen Chiou, PhD, Department of Life Science, Graduate Institute of Biotechnology, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Road, Hualien 97401, Taiwan, R.O.C.
| |
Collapse
|
15
|
Martin LJ, Akhavan B, Bilek MMM. Electric fields control the orientation of peptides irreversibly immobilized on radical-functionalized surfaces. Nat Commun 2018; 9:357. [PMID: 29367659 PMCID: PMC5783936 DOI: 10.1038/s41467-017-02545-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/07/2017] [Indexed: 01/12/2023] Open
Abstract
Surface functionalization of an implantable device with bioactive molecules can overcome adverse biological responses by promoting specific local tissue integration. Bioactive peptides have advantages over larger protein molecules due to their robustness and sterilizability. Their relatively small size presents opportunities to control the peptide orientation on approach to a surface to achieve favourable presentation of bioactive motifs. Here we demonstrate control of the orientation of surface-bound peptides by tuning electric fields at the surface during immobilization. Guided by computational simulations, a peptide with a linear conformation in solution is designed. Electric fields are used to control the peptide approach towards a radical-functionalized surface. Spontaneous, irreversible immobilization is achieved when the peptide makes contact with the surface. Our findings show that control of both peptide orientation and surface concentration is achieved simply by varying the solution pH or by applying an electric field as delivered by a small battery.
Collapse
Affiliation(s)
- Lewis J Martin
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
| | - Behnam Akhavan
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia.
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Marcela M M Bilek
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia.
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia.
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
16
|
Zhang YS, Zhu C, Xia Y. Inverse Opal Scaffolds and Their Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:10.1002/adma.201701115. [PMID: 28649794 PMCID: PMC5581229 DOI: 10.1002/adma.201701115] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 03/23/2017] [Indexed: 05/04/2023]
Abstract
Three-dimensional porous scaffolds play a pivotal role in tissue engineering and regenerative medicine by functioning as biomimetic substrates to manipulate cellular behaviors. While many techniques have been developed to fabricate porous scaffolds, most of them rely on stochastic processes that typically result in scaffolds with pores uncontrolled in terms of size, structure, and interconnectivity, greatly limiting their use in tissue regeneration. Inverse opal scaffolds, in contrast, possess uniform pores inheriting from the template comprised of a closely packed lattice of monodispersed microspheres. The key parameters of such scaffolds, including architecture, pore structure, porosity, and interconnectivity, can all be made uniform across the same sample and among different samples. In conjunction with a tight control over pore sizes, inverse opal scaffolds have found widespread use in biomedical applications. In this review, we provide a detailed discussion on this new class of advanced materials. After a brief introduction to their history and fabrication, we highlight the unique advantages of inverse opal scaffolds over their non-uniform counterparts. We then showcase their broad applications in tissue engineering and regenerative medicine, followed by a summary and perspective on future directions.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Chunlei Zhu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- School of Chemistry and Biochemistry, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
17
|
Kuo YC, Rajesh R. Guided differentiation and tissue regeneration of induced pluripotent stem cells using biomaterials. J Taiwan Inst Chem Eng 2017. [DOI: 10.1016/j.jtice.2017.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Kuo YC, Lin CY, Li JS, Lou YI. Wheat germ agglutinin-conjugated liposomes incorporated with cardiolipin to improve neuronal survival in Alzheimer's disease treatment. Int J Nanomedicine 2017; 12:1757-1774. [PMID: 28280340 PMCID: PMC5340244 DOI: 10.2147/ijn.s128396] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Curcumin (CRM) and nerve growth factor (NGF) were entrapped in liposomes (LIP) with surface wheat germ agglutinin (WGA) to downregulate the phosphorylation of kinases in Alzheimer’s disease (AD) therapy. Cardiolipin (CL)-conjugated LIP carrying CRM (CRM-CL/LIP) and also carrying NGF (NGF-CL/LIP) were used with AD models of SK-N-MC cells and Wistar rats after an insult with β-amyloid peptide (Aβ). We found that CRM-CL/LIP inhibited the expression of phosphorylated p38 (p-p38), phosphorylated c-Jun N-terminal kinase (p-JNK), and p-tau protein at serine 202 and prevented neurodegeneration of SK-N-MC cells. In addition, NGF-CL/LIP could enhance the quantities of p-neurotrophic tyrosine kinase receptor type 1 and p-extracellular signal-regulated kinase 5 for neuronal rescue. Moreover, WGA-grafted CRM-CL/LIP and WGA-grafted NGF-CL/LIP significantly improved the permeation of CRM and NGF across the blood–brain barrier, reduced Aβ plaque deposition and the malondialdehyde level, and increased the percentage of normal neurons and cholinergic activity in the hippocampus of AD rats. Based on the marker expressions and in vivo evidence, current LIP carriers can be promising drug delivery systems to protect nervous tissue against Aβ-induced apoptosis in the brain during the clinical management of AD.
Collapse
Affiliation(s)
| | | | - Jay-Shake Li
- Department of Psychology, National Chung Cheng University, Chia-Yi
| | - Yung-I Lou
- Department of Accounting, Providence University, Taichung, Taiwan, Republic of China
| |
Collapse
|
19
|
Wang PY, Thissen H, Kingshott P. Modulation of human multipotent and pluripotent stem cells using surface nanotopographies and surface-immobilised bioactive signals: A review. Acta Biomater 2016; 45:31-59. [PMID: 27596488 DOI: 10.1016/j.actbio.2016.08.054] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/30/2016] [Accepted: 08/30/2016] [Indexed: 02/08/2023]
Abstract
The ability to control the interactions of stem cells with synthetic surfaces is proving to be effective and essential for the quality of passaged stem cells and ultimately the success of regenerative medicine. The stem cell niche is crucial for stem cell self-renewal and differentiation. Thus, mimicking the stem cell niche, and here in particular the extracellular matrix (ECM), in vitro is an important goal for the expansion of stem cells and their applications. Here, surface nanotopographies and surface-immobilised biosignals have been identified as major factors that control stem cell responses. The development of tailored surfaces having an optimum nanotopography and displaying suitable biosignals is proposed to be essential for future stem cell culture, cell therapy and regenerative medicine applications. While early research in the field has been restricted by the limited availability of micro- and nanofabrication techniques, new approaches involving the use of advanced fabrication and surface immobilisation methods are starting to emerge. In addition, new cell types such as induced pluripotent stem cells (iPSCs) have become available in the last decade, but have not been fully understood. This review summarises significant advances in the area and focuses on the approaches that are aimed at controlling the behavior of human stem cells including maintenance of their self-renewal ability and improvement of their lineage commitment using nanotopographies and biosignals. More specifically, we discuss developments in biointerface science that are an important driving force for new biomedical materials and advances in bioengineering aiming at improving stem cell culture protocols and 3D scaffolds for clinical applications. Cellular responses revolve around the interplay between the surface properties of the cell culture substrate and the biomolecular composition of the cell culture medium. Determination of the precise role played by each factor, as well as the synergistic effects amongst the factors, all of which influence stem cell responses is essential for future developments. This review provides an overview of the current state-of-the-art in the design of complex material surfaces aimed at being the next generation of tools tailored for applications in cell culture and regenerative medicine. STATEMENT OF SIGNIFICANCE This review focuses on the effect of surface nanotopographies and surface-bound biosignals on human stem cells. Recently, stem cell research attracts much attention especially the induced pluripotent stem cells (iPSCs) and direct lineage reprogramming. The fast advance of stem cell research benefits disease treatment and cell therapy. On the other hand, surface property of cell adhered materials has been demonstrated very important for in vitro cell culture and regenerative medicine. Modulation of cell behavior using surfaces is costeffective and more defined. Thus, we summarise the recent progress of modulation of human stem cells using surface science. We believe that this review will capture a broad audience interested in topographical and chemical patterning aimed at understanding complex cellular responses to biomaterials.
Collapse
|
20
|
Regeneration of neurite-like cells from induced pluripotent stem cells in self-assembled hyaluronic acid-gelatin microhydrogel. J Taiwan Inst Chem Eng 2016. [DOI: 10.1016/j.jtice.2016.07.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
21
|
Chen C, Kong X, Lee IS. Modification of surface/neuron interfaces for neural cell-type specific responses: a review. ACTA ACUST UNITED AC 2015; 11:014108. [PMID: 26694886 DOI: 10.1088/1748-6041/11/1/014108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Surface/neuron interfaces have played an important role in neural repair including neural prostheses and tissue engineered scaffolds. This comprehensive literature review covers recent studies on the modification of surface/neuron interfaces. These interfaces are identified in cases both where the surfaces of substrates or scaffolds were in direct contact with cells and where the surfaces were modified to facilitate cell adhesion and controlling cell-type specific responses. Different sources of cells for neural repair are described, such as pheochromocytoma neuronal-like cell, neural stem cell (NSC), embryonic stem cell (ESC), mesenchymal stem cell (MSC) and induced pluripotent stem cell (iPS). Commonly modified methods are discussed including patterned surfaces at micro- or nano-scale, surface modification with conducting coatings, and functionalized surfaces with immobilized bioactive molecules. These approaches to control cell-type specific responses have enormous potential implications in neural repair.
Collapse
Affiliation(s)
- Cen Chen
- Bio-X Center, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | | | | |
Collapse
|
22
|
Zomer HD, Vidane AS, Gonçalves NN, Ambrósio CE. Mesenchymal and induced pluripotent stem cells: general insights and clinical perspectives. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:125-34. [PMID: 26451119 PMCID: PMC4592031 DOI: 10.2147/sccaa.s88036] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells have awakened a great deal of interest in regenerative medicine due to their plasticity, and immunomodulatory and anti-inflammatory properties. They are high-yield and can be acquired through noninvasive methods from adult tissues. Moreover, they are nontumorigenic and are the most widely studied. On the other hand, induced pluripotent stem (iPS) cells can be derived directly from adult cells through gene reprogramming. The new iPS technology avoids the embryo destruction or manipulation to generate pluripotent cells, therefore, are exempt from ethical implication surrounding embryonic stem cell use. The pre-differentiation of iPS cells ensures the safety of future approaches. Both mesenchymal stem cells and iPS cells can be used for autologous cell transplantations without the risk of immune rejection and represent a great opportunity for future alternative therapies. In this review we discussed the therapeutic perspectives using mesenchymal and iPS cells.
Collapse
Affiliation(s)
- Helena D Zomer
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Atanásio S Vidane
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Natalia N Gonçalves
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Carlos E Ambrósio
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| |
Collapse
|
23
|
Antunes JC, Tsaryk R, Gonçalves RM, Pereira CL, Landes C, Brochhausen C, Ghanaati S, Barbosa MA, Kirkpatrick CJ. Poly(γ-Glutamic Acid) as an Exogenous Promoter of Chondrogenic Differentiation of Human Mesenchymal Stem/Stromal Cells. Tissue Eng Part A 2015; 21:1869-85. [PMID: 25760236 DOI: 10.1089/ten.tea.2014.0386] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cartilage damage and/or aging effects can cause constant pain, which limits the patient's quality of life. Although different strategies have been proposed to enhance the limited regenerative capacity of cartilage tissue, the full production of native and functional cartilaginous extracellular matrix (ECM) has not yet been achieved. Poly(γ-glutamic acid) (γ-PGA), a naturally occurring polyamino acid, biodegradable into glutamate residues, has been explored for tissue regeneration. In this work, γ-PGA's ability to support the production of cartilaginous ECM by human bone marrow mesenchymal stem/stromal cells (MSCs) and nasal chondrocytes (NCs) was investigated. MSC and NC pellets were cultured in basal medium (BM), chondrogenic medium (CM), and CM-γ-PGA-supplemented medium (CM+γ-PGA) over a period of 21 days. Pellet size/shape was monitored with time. At 14 and 21 days of culture, the presence of sulfated glycosaminoglycans (sGAGs), type II collagen (Col II), Sox-9, aggrecan, type XI collagen (Col XI), type X collagen (Col X), calcium deposits, and type I collagen (Col I) was analyzed. After excluding γ-PGA's cytotoxicity, earlier cell condensation, higher sGAG content, Col II, Sox-9 (day 14), aggrecan, and Col X (day 14) production was observed in γ-PGA-supplemented MSC cultures, with no signs of mineralization or Col I. These effects were not evident with NCs. However, Sox-9 (at day 14) and Col X (at days 14 and 21) were increased, decreased, or absent, respectively. Overall, γ-PGA improved chondrogenic differentiation of MSCs, increasing ECM production earlier in culture. It is proposed that γ-PGA incorporation in novel biomaterials has a beneficial impact on future approaches for cartilage regeneration.
Collapse
Affiliation(s)
- Joana C Antunes
- 1Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,3Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Roman Tsaryk
- 3Faculdade de Engenharia, Universidade do Porto, Porto, Portugal.,4Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Raquel M Gonçalves
- 1Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Catarina Leite Pereira
- 1Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,5ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Constantin Landes
- 6Department of Oral, Cranio-Maxillofacial and Facial Plastic Surgery, University Medical Center of the Goethe University, Frankfurt am Main, Germany.,7Sana Hospital Offenbach, Offenbach, Germany
| | - Christoph Brochhausen
- 8REPAIR Lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Shahram Ghanaati
- 6Department of Oral, Cranio-Maxillofacial and Facial Plastic Surgery, University Medical Center of the Goethe University, Frankfurt am Main, Germany.,7Sana Hospital Offenbach, Offenbach, Germany.,8REPAIR Lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Mário A Barbosa
- 1Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,5ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - C James Kirkpatrick
- 8REPAIR Lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
24
|
Tong Z, Solanki A, Hamilos A, Levy O, Wen K, Yin X, Karp JM. Application of biomaterials to advance induced pluripotent stem cell research and therapy. EMBO J 2015; 34:987-1008. [PMID: 25766254 PMCID: PMC4406648 DOI: 10.15252/embj.201490756] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/25/2015] [Accepted: 02/17/2015] [Indexed: 12/19/2022] Open
Abstract
Derived from any somatic cell type and possessing unlimited self-renewal and differentiation potential, induced pluripotent stem cells (iPSCs) are poised to revolutionize stem cell biology and regenerative medicine research, bringing unprecedented opportunities for treating debilitating human diseases. To overcome the limitations associated with safety, efficiency, and scalability of traditional iPSC derivation, expansion, and differentiation protocols, biomaterials have recently been considered. Beyond addressing these limitations, the integration of biomaterials with existing iPSC culture platforms could offer additional opportunities to better probe the biology and control the behavior of iPSCs or their progeny in vitro and in vivo. Herein, we discuss the impact of biomaterials on the iPSC field, from derivation to tissue regeneration and modeling. Although still exploratory, we envision the emerging combination of biomaterials and iPSCs will be critical in the successful application of iPSCs and their progeny for research and clinical translation.
Collapse
Affiliation(s)
- Zhixiang Tong
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Aniruddh Solanki
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Allison Hamilos
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Oren Levy
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Kendall Wen
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Xiaolei Yin
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Jeffrey M Karp
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| |
Collapse
|
25
|
Shi L, Yang N, Zhang H, Chen L, Tao L, Wei Y, Liu H, Luo Y. A novel poly(γ-glutamic acid)/silk-sericin hydrogel for wound dressing: Synthesis, characterization and biological evaluation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 48:533-40. [DOI: 10.1016/j.msec.2014.12.047] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 11/05/2014] [Accepted: 12/09/2014] [Indexed: 11/29/2022]
|
26
|
Wang CC, Kuo YC. Capillary electrophoresis of induced pluripotent stem cells during differentiation toward neurons. J Taiwan Inst Chem Eng 2014. [DOI: 10.1016/j.jtice.2014.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
27
|
João CFC, Vasconcelos JM, Silva JC, Borges JP. An overview of inverted colloidal crystal systems for tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:437-54. [PMID: 24328724 DOI: 10.1089/ten.teb.2013.0402] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Scaffolding is at the heart of tissue engineering but the number of techniques available for turning biomaterials into scaffolds displaying the features required for a tissue engineering application is somewhat limited. Inverted colloidal crystals (ICCs) are inverse replicas of an ordered array of monodisperse colloidal particles, which organize themselves in packed long-range crystals. The literature on ICC systems has grown enormously in the past 20 years, driven by the need to find organized macroporous structures. Although replicating the structure of packed colloidal crystals (CCs) into solid structures has produced a wide range of advanced materials (e.g., photonic crystals, catalysts, and membranes) only in recent years have ICCs been evaluated as devices for medical/pharmaceutical and tissue engineering applications. The geometry, size, pore density, and interconnectivity are features of the scaffold that strongly affect the cell environment with consequences on cell adhesion, proliferation, and differentiation. ICC scaffolds are highly geometrically ordered structures with increased porosity and connectivity, which enhances oxygen and nutrient diffusion, providing optimum cellular development. In comparison to other types of scaffolds, ICCs have three major unique features: the isotropic three-dimensional environment, comprising highly uniform and size-controllable pores, and the presence of windows connecting adjacent pores. Thus far, this is the only technique that guarantees these features with a long-range order, between a few nanometers and thousands of micrometers. In this review, we present the current development status of ICC scaffolds for tissue engineering applications.
Collapse
Affiliation(s)
- Carlos Filipe C João
- 1 CENIMAT/I3N, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa , Caparica, Portugal
| | | | | | | |
Collapse
|
28
|
Neural differentiation of pluripotent cells in 3D alginate-based cultures. Biomaterials 2014; 35:4636-45. [PMID: 24631250 DOI: 10.1016/j.biomaterials.2014.02.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 02/21/2014] [Indexed: 12/14/2022]
Abstract
Biomaterial-supported culture methods, allowing for directed three-dimensional differentiation of stem cells are an alternative to canonical two-dimensional cell cultures. In this paper, we evaluate the suitability of alginate for three-dimensional cultures to enhance differentiation of mouse embryonic stem cells (mESCs) towards neural lineages. We tested whether encapsulation of mESCs within alginate beads could support and/or enhance neural differentiation with respect to two-dimensional cultures. We encapsulated cells in beads of alginate with or without modification by fibronectin (Fn) or hyaluronic acid (HA). Gene expression analysis showed that cells grown in alginate and alginate-HA present increased differentiation toward neural lineages with respect to the two-dimensional control and to Fn group. Immunocytochemistry analyses confirmed these results, further showing terminal differentiation of neurons as seen by the expression of synaptic markers and markers of different neuronal subtypes. Our data show that alginate, alone or modified, is a suitable biomaterial to promote in vitro differentiation of pluripotent cells toward neural fates.
Collapse
|
29
|
Joddar B, Ito Y. Artificial niche substrates for embryonic and induced pluripotent stem cell cultures. J Biotechnol 2013; 168:218-28. [DOI: 10.1016/j.jbiotec.2013.04.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/13/2013] [Accepted: 04/29/2013] [Indexed: 01/27/2023]
|
30
|
Chung CY, Yang JT, Kuo YC. Polybutylcyanoacrylate nanoparticles for delivering hormone response element-conjugated neurotrophin-3 to the brain of intracerebral hemorrhagic rats. Biomaterials 2013; 34:9717-27. [PMID: 24034503 DOI: 10.1016/j.biomaterials.2013.08.083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 08/27/2013] [Indexed: 01/09/2023]
Abstract
Hypertensive intracerebral hemorrhage (ICH) is a rapidly evolutional pathology, inducing necrotic cell death followed by apoptosis, and alters gene expression levels in surrounding tissue of an injured brain. For ICH therapy by controlled gene release, the development of intravenously administrable delivery vectors to promote the penetration across the blood-brain barrier (BBB) is a critical challenge. To enhance transfer efficiency of genetic materials under hypoxic conditions, polybutylcyanoacrylate (PBCA) nanoparticles (NPs) were used to mediate the intracellular transport of plasmid neurotrophin-3 (NT-3) containing hormone response element (HRE) with a cytomegalovirus (cmv) promoter and to differentiate induced pluripotent stem cells (iPSCs). The differentiation ability of iPSCs to neurons was justified by various immunological stains for protein fluorescence. The effect of PBCA NP/cmvNT-3-HRE complexes on treating ICH rats was studied by immunostaining, western blotting and Nissl staining. We found that the treatments with PBCA NP/cmvNT-3-HRE complexes increased the capability of differentiating iPSCs to express NT-3, TrkC and MAP-2. Moreover, PBCA NPs could protect cmvNT-3-HRE against degradation with EcoRI/PstI and DNase I in vitro and raise the delivery across the BBB in vivo. The administration of PBCA NP/cmvNT-3-HRE complexes increased the expression of NT-3, inhibited the expression of apoptosis-inducing factor, cleaved caspase-3 and DNA fragmentation, and reduced the cell death rate after ICH in vivo. PBCA NPs are demonstrated as an appropriate delivery system for carrying cmvNT-3-HRE to the brain for ICH therapy.
Collapse
Affiliation(s)
- Chiu-Yen Chung
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | | | | |
Collapse
|
31
|
Chung CY, Yang JT, Kuo YC. Polybutylcyanoacrylate nanoparticle-mediated neurotrophin-3 gene delivery for differentiating iPS cells into neurons. Biomaterials 2013; 34:5562-70. [PMID: 23623427 DOI: 10.1016/j.biomaterials.2013.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/03/2013] [Indexed: 12/12/2022]
Abstract
Guided neuronal differentiation of induced pluripotent stem cells (iPSCs) with genetic regulation is an important issue in biomedical research and in clinical practice for nervous regeneration and repair. To enhance the intracellular delivery of plasmid DNA (pDNA), polybutylcyanoacrylate (PBCA) nanoparticles (NPs) were employed to mediate the transport of neurotrophin-3 (NT-3) into iPSCs. The ability of iPSCs to differentiate into neuronal lineages was shown by immunofluorescent staining, western blotting, and flow cytometry. By transmission electron microscopy, we found that PBCA NPs could efficiently grasp pDNA, thereby increasing the particle size and conferring a negative surface charge. In addition, the treatments with PBCA NP/NT-3 complexes enhanced the expression of NT-3, TrkC, NH-H, NSE, and PSD95 by differentiating iPSCs. Neurons produced from iPSCs were incapable of returning to pluripotency, demonstrating with a series of differentiation scheme for adipogenesis and osteogenesis. The pretreatment with PBCA NP/NT-3 complexes can be one of critical biotechnologies and effective delivery systems in gene transfection to accelerate the differentiation of iPSCs into neurons.
Collapse
Affiliation(s)
- Chiu-Yen Chung
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | | | | |
Collapse
|
32
|
Kuo YC, Wang CC. Guided differentiation of induced pluripotent stem cells into neuronal lineage in alginate–chitosan–gelatin hydrogels with surface neuron growth factor. Colloids Surf B Biointerfaces 2013; 104:194-9. [DOI: 10.1016/j.colsurfb.2013.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 12/28/2012] [Accepted: 01/01/2013] [Indexed: 10/27/2022]
|
33
|
Alginate-Based Biomaterials for Regenerative Medicine Applications. MATERIALS 2013; 6:1285-1309. [PMID: 28809210 PMCID: PMC5452316 DOI: 10.3390/ma6041285] [Citation(s) in RCA: 715] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 02/19/2013] [Accepted: 03/19/2013] [Indexed: 02/07/2023]
Abstract
Alginate is a natural polysaccharide exhibiting excellent biocompatibility and biodegradability, having many different applications in the field of biomedicine. Alginate is readily processable for applicable three-dimensional scaffolding materials such as hydrogels, microspheres, microcapsules, sponges, foams and fibers. Alginate-based biomaterials can be utilized as drug delivery systems and cell carriers for tissue engineering. Alginate can be easily modified via chemical and physical reactions to obtain derivatives having various structures, properties, functions and applications. Tuning the structure and properties such as biodegradability, mechanical strength, gelation property and cell affinity can be achieved through combination with other biomaterials, immobilization of specific ligands such as peptide and sugar molecules, and physical or chemical crosslinking. This review focuses on recent advances in the use of alginate and its derivatives in the field of biomedical applications, including wound healing, cartilage repair, bone regeneration and drug delivery, which have potential in tissue regeneration applications.
Collapse
|
34
|
Kuo YC, Lin CC. Accelerated nerve regeneration using induced pluripotent stem cells in chitin–chitosan–gelatin scaffolds with inverted colloidal crystal geometry. Colloids Surf B Biointerfaces 2013; 103:595-600. [DOI: 10.1016/j.colsurfb.2012.11.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 10/31/2012] [Accepted: 11/02/2012] [Indexed: 01/22/2023]
|