201
|
Lee JK, Choi IS, Oh TI, Lee E. Cell-Surface Engineering for Advanced Cell Therapy. Chemistry 2018; 24:15725-15743. [PMID: 29791047 DOI: 10.1002/chem.201801710] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/22/2018] [Indexed: 12/16/2022]
Abstract
Stem cells opened great opportunity to overcome diseases that conventional therapy had only limited success. Use of scaffolds made from biomaterials not only helps handling of stem cells for delivery or transplantation but also supports enhanced cell survival. Likewise, cell encapsulation can provide stability for living animal cells even in a state of separateness. Although various chemical reactions were tried to encapsulate stolid microbial cells such as yeasts, a culture environment for the growth of animal cells allows only highly biocompatible reactions. Therefore, the animal cells were mostly encapsulated in hydrogels, which resulted in enhanced cell survival. Interestingly, major findings of chemistry on biological interfaces demonstrate that cell encapsulation in hydrogels have a further a competence for modulating cell characteristics that can go beyond just enhancing the cell survival. In this review, we present a comprehensive overview on the chemical reactions applied to hydrogel-based cell encapsulation and their effects on the characteristics and behavior of living animal cells.
Collapse
Affiliation(s)
- Jungkyu K Lee
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Korea
| | - Insung S Choi
- Department of Chemistry and Center for Cell-Encapsulation Research, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Tong In Oh
- Department of Biomedical Engineering, Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - EunAh Lee
- Impedance Imaging Research Center (IIRC), Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| |
Collapse
|
202
|
Lueckgen A, Garske DS, Ellinghaus A, Desai RM, Stafford AG, Mooney DJ, Duda GN, Cipitria A. Hydrolytically-degradable click-crosslinked alginate hydrogels. Biomaterials 2018; 181:189-198. [PMID: 30086448 DOI: 10.1016/j.biomaterials.2018.07.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 12/30/2022]
Abstract
Degradable biomaterials aim to recapitulate the dynamic microenvironment that cells are naturally exposed to. By oxidizing the alginate polymer backbone, thereby rendering it susceptible to hydrolysis, and crosslinking it via norbornene-tetrazine click chemistry, we can control rheological, mechanical, and degradation properties of resulting hydrogels. Chemical modifications were confirmed by nuclear magnetic resonance (NMR) and the resulting mechanical properties measured by rheology and unconfined compression testing, demonstrating that these are both a function of norbornene coupling and oxidation state. The degradation behavior was verified by tracking mechanical and swelling behavior over time, showing that degradation could be decoupled from initial mechanical properties. The cell compatibility was assessed in 2D and 3D using a mouse pre-osteoblast cell line and testing morphology, proliferation, and viability. Cells attached, spread and proliferated in 2D and retained a round morphology and stable number in 3D, while maintaining high viability in both contexts over 7 days. Finally, oxidized and unoxidized control materials were implanted subcutaneously into the backs of C57/Bl6 mice, and recovered after 8 weeks. Histological staining revealed morphological differences and fibrous tissue infiltration only in oxidized materials. These materials with tunable and decoupled mechanical and degradation behavior could be useful in many tissue engineering applications.
Collapse
Affiliation(s)
- Aline Lueckgen
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Daniela S Garske
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Rajiv M Desai
- School of Engineering and Applied Sciences - Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Alexander G Stafford
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - David J Mooney
- School of Engineering and Applied Sciences - Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Georg N Duda
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Amaia Cipitria
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, 14476 Potsdam, Germany.
| |
Collapse
|
203
|
Yu Z, Zhou Z, Huang G, Zheng X, Wu L, Zhao S, Meng F. Two-Dimensional FTIR Spectroscopic Characterization of Functional Groups of NaOCl-Exposed Alginate: Insights into Membrane Refouling after Online Chemical Cleaning. ACS APPLIED BIO MATERIALS 2018; 1:593-603. [DOI: 10.1021/acsabm.8b00082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Zhong Yu
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, PR China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhongbo Zhou
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, PR China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Guocheng Huang
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, PR China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Xing Zheng
- Department of Civil and Environmental Engineering, Xi’an University of Technology, Xi’an 710048, China
| | - Linjie Wu
- Department of Civil and Environmental Engineering, Xi’an University of Technology, Xi’an 710048, China
| | - Shanshan Zhao
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, PR China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Fangang Meng
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, PR China
- Guangdong Provincial Key Laboratory of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
204
|
Ali A, Ahmed S. Recent Advances in Edible Polymer Based Hydrogels as a Sustainable Alternative to Conventional Polymers. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6940-6967. [PMID: 29878765 DOI: 10.1021/acs.jafc.8b01052] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The over increasing demand of eco-friendly materials to counter various problems, such as environmental issues, economics, sustainability, biodegradability, and biocompatibility, open up new fields of research highly focusing on nature-based products. Edible polymer based materials mainly consisting of polysaccharides, proteins, and lipids could be a prospective contender to handle such problems. Hydrogels based on edible polymer offer many valuable properties compared to their synthetic counterparts. Edible polymers can contribute to the reduction of environmental contamination, advance recyclability, provide sustainability, and thereby increase its applicability along with providing environmentally benign products. This review is highly emphasizing on toward the development of hydrogels from edible polymer, their classification, properties, chemical modification, and their potential applications. The application of edible polymer hydrogels covers many areas including the food industry, agricultural applications, drug delivery to tissue engineering in the biomedical field and provide more safe and attractive products in the pharmaceutical, agricultural, and environmental fields, etc.
Collapse
Affiliation(s)
- Akbar Ali
- Department of Chemistry , Jamia Millia Islamia , New Delhi , 110025 , India
| | - Shakeel Ahmed
- Department of Chemistry , Government Degree College Mendhar , Jammu , Jammu and Kashmir , 185211 , India
- Higher Education Department , Government of Jammu and Kashmir , Jammu , 180001 , India
| |
Collapse
|
205
|
Hettiaratchi MH, Schudel A, Rouse T, García AJ, Thomas SN, Guldberg RE, McDevitt TC. A rapid method for determining protein diffusion through hydrogels for regenerative medicine applications. APL Bioeng 2018; 2:026110. [PMID: 31069307 PMCID: PMC6324205 DOI: 10.1063/1.4999925] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 04/30/2018] [Indexed: 12/24/2022] Open
Abstract
Hydrogels present versatile platforms for the encapsulation and delivery of proteins and cells for regenerative medicine applications. However, differences in hydrogel cross-linking density, polymer weight content, and affinity for proteins all contribute to diverse diffusion rates of proteins through hydrogel networks. Here, we describe a simple method to accurately measure protein diffusion through hydrogels, within a few hours and without the use of large amounts of protein. We tracked the diffusion of several proteins of varying molecular weights along the axial direction of capillary tubes filled with alginate, collagen, or poly(ethylene glycol) hydrogels. The rate of protein diffusion decreased with increasing molecular weight. A computational model of protein diffusion through capillary tubes was also created to predict and verify experimental protein diffusion coefficients. This in vitro capillary tube-based method of measuring protein diffusion represents a simple strategy to interrogate protein diffusion through natural and synthetic hydrogels and aid in the design of better biomaterial-based delivery vehicles that can effectively modulate protein release.
Collapse
Affiliation(s)
- Marian H. Hettiaratchi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive NW, Atlanta, Georgia 30332, USA
| | | | | | | | | | | | | |
Collapse
|
206
|
Miao T, Wang J, Zeng Y, Liu G, Chen X. Polysaccharide-Based Controlled Release Systems for Therapeutics Delivery and Tissue Engineering: From Bench to Bedside. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700513. [PMID: 29721408 PMCID: PMC5908359 DOI: 10.1002/advs.201700513] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/19/2017] [Indexed: 05/08/2023]
Abstract
Polysaccharides or polymeric carbohydrate molecules are long chains of monosaccharides that are linked by glycosidic bonds. The naturally based structural materials are widely applied in biomedical applications. This article covers four different types of polysaccharides (i.e., alginate, chitosan, hyaluronic acid, and dextran) and emphasizes their chemical modification, preparation approaches, preclinical studies, and clinical translations. Different cargo fabrication techniques are also presented in the third section. Recent progresses in preclinical applications are then discussed, including tissue engineering and treatment of diseases in both therapeutic and monitoring aspects. Finally, clinical translational studies with ongoing clinical trials are summarized and reviewed. The promise of new development in nanotechnology and polysaccharide chemistry helps clinical translation of polysaccharide-based drug delivery systems.
Collapse
Affiliation(s)
- Tianxin Miao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- Collaborative Innovation Center of Guangxi Biological Medicine and theMedical and Scientific Research CenterGuangxi Medical UniversityNanning530021China
| | - Yun Zeng
- Department of PharmacologyXiamen Medical CollegeXiamen361008China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell BiologySchool of Life SciencesXiamen UniversityXiamen361102China
- State Key Laboratory of Physical Chemistry of Solid Surfaces and The MOE Key Laboratory of Spectrochemical Analysis & InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and NanomedicineNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
207
|
Pandey N, Hakamivala A, Xu C, Hariharan P, Radionov B, Huang Z, Liao J, Tang L, Zimmern P, Nguyen KT, Hong Y. Biodegradable Nanoparticles Enhanced Adhesiveness of Mussel-Like Hydrogels at Tissue Interface. Adv Healthc Mater 2018; 7:e1701069. [PMID: 29205950 DOI: 10.1002/adhm.201701069] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Indexed: 12/29/2022]
Abstract
Popular bioadhesives, such as fibrin, cyanoacrylate, and albumin-glutaraldehyde based materials, have been applied for clinical applications in wound healing, drug delivery, and bone and soft tissue engineering; however, their performances are limited by weak adhesion strength and rapid degradation. In this study a mussel-inspired, nanocomposite-based, biodegradable tissue adhesive is developed by blending poly(lactic-co-glycolic acid) (PLGA) or N-hydroxysuccinimide modified PLGA nanoparticles (PLGA-NHS) with mussel-inspired alginate-dopamine polymer (Alg-Dopa). Adhesive strength measurement of the nanocomposites on porcine skin-muscle constructs reveals that the incorporation of nanoparticles in Alg-Dopa significantly enhances the tissue adhesive strength compared to the mussel-inspired adhesive alone. The nanocomposite formed by PLGA-NHS nanoparticles shows higher lap shear strength of 33 ± 3 kPa, compared to that of Alg-Dopa hydrogel alone (14 ± 2 kPa). In addition, these nanocomposites are degradable and cytocompatible in vitro, and elicit in vivo minimal inflammatory responses in a rat model, suggesting clinical potential of these nanocomposites as bioadhesives.
Collapse
Affiliation(s)
- Nikhil Pandey
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Amirhossein Hakamivala
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Cancan Xu
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Prashant Hariharan
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Boris Radionov
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Zhong Huang
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
| | - Jun Liao
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Liping Tang
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Philippe Zimmern
- Department of Urology University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Kytai T. Nguyen
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| | - Yi Hong
- Department of Bioengineering University of Texas at Arlington Arlington TX 76010 USA
- Joint Biomedical Engineering Program University of Texas Southwestern Medical Center Dallas TX 75390 USA
| |
Collapse
|
208
|
Madrigal JL, Sharma SN, Campbell KT, Stilhano RS, Gijsbers R, Silva EA. Microgels produced using microfluidic on-chip polymer blending for controlled released of VEGF encoding lentivectors. Acta Biomater 2018; 69:265-276. [PMID: 29398644 PMCID: PMC6819130 DOI: 10.1016/j.actbio.2018.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/01/2022]
Abstract
Alginate hydrogels are widely used as delivery vehicles due to their ability to encapsulate and release a wide range of cargos in a gentle and biocompatible manner. The release of encapsulated therapeutic cargos can be promoted or stunted by adjusting the hydrogel physiochemical properties. However, the release from such systems is often skewed towards burst-release or lengthy retention. To address this, we hypothesized that the overall magnitude of burst release could be adjusted by combining microgels with distinct properties and release behavior. Microgel suspensions were generated using a process we have termed on-chip polymer blending to yield composite suspensions of a range of microgel formulations. In this manner, we studied how alginate percentage and degradation relate to the release of lentivectors. Whereas changes in alginate percentage had a minimal impact on lentivector release, microgel degradation led to a 3-fold increase, and near complete release, over 10 days. Furthermore, by controlling the amount of degradable alginate present within microgels the relative rate of release can be adjusted. A degradable formulation of microgels was used to deliver vascular endothelial growth factor (VEGF)-encoding lentivectors in the chick chorioallantoic membrane (CAM) assay and yielded a proangiogenic response in comparison to the same lentivectors delivered in suspension. The utility of blended microgel suspensions may provide an especially appealing platform for the delivery of lentivectors or similarly sized therapeutics. STATEMENT OF SIGNIFICANCE Genetic therapeutics hold considerable potential for the treatment of diseases and disorders including ischemic cardiovascular diseases. To realize this potential, genetic vectors must be precisely and efficiently delivered to targeted regions of the body. However, conventional methods of delivery do not provide sufficient spatial and temporal control. Here, we demonstrate how alginate microgels provide a basis for developing systems for controlled genetic vector release. We adjust the physiochemical properties of alginate for quicker or slower release, and we demonstrate how combining distinct formulations of microgels can tune the release of the overall composite microgel suspension. These composite suspensions are generated using a straightforward and powerful application of droplet microfluidics which allows for the real-time generation of a composite suspension.
Collapse
Affiliation(s)
- Justin L Madrigal
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Shonit N Sharma
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Kevin T Campbell
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Roberta S Stilhano
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Rik Gijsbers
- Department of Pharmaceutic and Pharmacological Sciences, Laboratory for Viral Vector Technology and Gene Therapy, KU Leuven-University of Leuven, Leuven, Belgium
| | - Eduardo A Silva
- Department of Biomedical Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
209
|
Synthesis and evaluation of anti-fungal activities of sodium alginate-amphotericin B conjugates. Int J Biol Macromol 2018; 108:1101-1109. [DOI: 10.1016/j.ijbiomac.2017.11.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 11/20/2022]
|
210
|
Torres A, Bidarra S, Pinto M, Aguiar P, Silva E, Barrias C. Guiding morphogenesis in cell-instructive microgels for therapeutic angiogenesis. Biomaterials 2018; 154:34-47. [DOI: 10.1016/j.biomaterials.2017.10.051] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022]
|
211
|
Jang J, Cha C. Multivalent Polyaspartamide Cross-Linker for Engineering Cell-Responsive Hydrogels with Degradation Behavior and Tunable Physical Properties. Biomacromolecules 2018; 19:691-700. [DOI: 10.1021/acs.biomac.8b00068] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Jinhyeong Jang
- Department of Chemistry, School of Natural Science and ‡School of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea
| | - Chaenyung Cha
- Department of Chemistry, School of Natural Science and ‡School of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea
| |
Collapse
|
212
|
Khoshzaban A, Keyhanvar P, Delrish E, Najafi F, Heidari Keshel S, Watanabe I, Valanezhad A, Jafarzadeh Kashi T. Alginate Microcapsules as Nutrient Suppliers: An In Vitro Study. CELL JOURNAL 2018; 20:25-30. [PMID: 29308615 PMCID: PMC5759677 DOI: 10.22074/cellj.2018.4508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/20/2017] [Indexed: 11/24/2022]
Abstract
Objective Alginate, known as a group of anionic polysaccharides extracted from seaweeds, has attracted the attention
of researchers because of its biocompatibility and degradability properties. Alginate has shown beneficial effects on
wound healing as it has similar function as extracellular matrix. Alginate microcapsules (AM) that are used in tissue
engineering as well as Dulbecco’s modified Eagle’s medium (DMEM) contain nutrients required for cell viability. The
purpose of this research was introducing AM in medium and nutrient reagent cells and making a comparison with
control group cells that have been normally cultured in long term.
Materials and Methods In this experimental study, AM were shaped in distilled water, it was dropped at 5 mL/hours
through a flat 25G5/8 sterile needle into a crosslinking bath containing 0.1 M calcium chloride to produce calcium
alginate microspheres. Then, the size of microcapsules (300-350 µm) were confirmed by Scanning Electron Microscopy
(SEM) images after the filtration for selection of the best size. Next, DMEM was injected into AM. Afterward, adipose-
derived mesenchymal stem cells (ADSCs) and Ringer’s serum were added. Then, MTT and DAPI assays were used
for cell viability and nucleus staining, respectively. Also, morphology of microcapsules was determined under invert
microscopy.
Results Evaluation of the cells performed for spatial media/microcapsules at the volume of 40 µl, showed ADSCs
after 1-day cell culture. Also, MTT assay results showed a significant difference in the viability of sustained-release
media injected to microcapsules (P<0.05). DAPI staining revealed living cells on the microcapsules after 1 to 7-day cell
culture.
Conclusion According to the results, AM had a positive effect on cell viability in scaffolds and tissue engineering and
provide nutrients needed in cell therapy.
Collapse
Affiliation(s)
- Ahad Khoshzaban
- Iranian Tissue Bank and Research Center, Imam Khomeini Medical Complex Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Stem Cell Preparation Unit, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Department of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Keyhanvar
- Stem Cell Preparation Unit, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Delrish
- Stem Cell Preparation Unit, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhood Najafi
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran
| | - Saeed Heidari Keshel
- Stem Cell Preparation Unit, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ikuya Watanabe
- Department of Bio-Engineering, Nagasaki University, Nagasaki, Japan
| | | | - Tahereh Jafarzadeh Kashi
- Iranian Tissue Bank and Research Center, Imam Khomeini Medical Complex Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Department of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran.Electronic Address:
| |
Collapse
|
213
|
Chen S, Cui S, Zhang H, Pei X, Hu J, Zhou Y, Liu Y. Cross-Linked Pectin Nanofibers with Enhanced Cell Adhesion. Biomacromolecules 2018; 19:490-498. [DOI: 10.1021/acs.biomac.7b01605] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Sainan Chen
- Key Laboratory of UV-Emitting Materials and Technology (Northeast Normal University), Ministry of Education, Changchun, Jilin 130024, P. R. China
| | - Sisi Cui
- School
of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P. R. China
| | - Hui Zhang
- Key Laboratory of UV-Emitting Materials and Technology (Northeast Normal University), Ministry of Education, Changchun, Jilin 130024, P. R. China
| | - Xuejing Pei
- School
of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P. R. China
| | - Junli Hu
- Key Laboratory of UV-Emitting Materials and Technology (Northeast Normal University), Ministry of Education, Changchun, Jilin 130024, P. R. China
| | - Yifa Zhou
- School
of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P. R. China
| | - Yichun Liu
- Key Laboratory of UV-Emitting Materials and Technology (Northeast Normal University), Ministry of Education, Changchun, Jilin 130024, P. R. China
| |
Collapse
|
214
|
Stanisci A, Aarstad OA, Tøndervik A, Sletta H, Dypås LB, Skjåk-Bræk G, Aachmann FL. Overall size of mannuronan C5-Epimerases influences their ability to epimerize modified alginates and alginate gels. Carbohydr Polym 2018; 180:256-263. [DOI: 10.1016/j.carbpol.2017.09.094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/11/2017] [Accepted: 09/27/2017] [Indexed: 10/18/2022]
|
215
|
Thomas D, O'Brien T, Pandit A. Toward Customized Extracellular Niche Engineering: Progress in Cell-Entrapment Technologies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1703948. [PMID: 29194781 DOI: 10.1002/adma.201703948] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/12/2017] [Indexed: 06/07/2023]
Abstract
The primary aim in tissue engineering is to repair, replace, and regenerate dysfunctional tissues to restore homeostasis. Cell delivery for repair and regeneration is gaining impetus with our understanding of constructing tissue-like environments. However, the perpetual challenge is to identify innovative materials or re-engineer natural materials to model cell-specific tissue-like 3D modules, which can seamlessly integrate and restore functions of the target organ. To devise an optimal functional microenvironment, it is essential to define how simple is complex enough to trigger tissue regeneration or restore cellular function. Here, the purposeful transition of cell immobilization from a cytoprotection point of view to that of a cell-instructive approach is examined, with advances in the understanding of cell-material interactions in a 3D context, and with a view to further application of the knowledge for the development of newer and complex hierarchical tissue assemblies for better examination of cell behavior and offering customized cell-based therapies for tissue engineering.
Collapse
Affiliation(s)
- Dilip Thomas
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
216
|
Alginate Utilization in Tissue Engineering and Cell Therapy. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2018. [DOI: 10.1007/978-981-10-6910-9_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
217
|
The effect of hydration on the material and mechanical properties of cellulose nanocrystal-alginate composites. Carbohydr Polym 2018; 179:186-195. [DOI: 10.1016/j.carbpol.2017.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/18/2017] [Accepted: 09/02/2017] [Indexed: 11/23/2022]
|
218
|
Sultan MT, Lee OJ, Kim SH, Ju HW, Park CH. Silk Fibroin in Wound Healing Process. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:115-126. [DOI: 10.1007/978-981-13-0947-2_7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
219
|
Reakasame S, Boccaccini AR. Oxidized Alginate-Based Hydrogels for Tissue Engineering Applications: A Review. Biomacromolecules 2017; 19:3-21. [DOI: 10.1021/acs.biomac.7b01331] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Supachai Reakasame
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| | - Aldo R. Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| |
Collapse
|
220
|
|
221
|
Ansari S, Diniz IM, Chen C, Sarrion P, Tamayol A, Wu BM, Moshaverinia A. Human Periodontal Ligament- and Gingiva-derived Mesenchymal Stem Cells Promote Nerve Regeneration When Encapsulated in Alginate/Hyaluronic Acid 3D Scaffold. Adv Healthc Mater 2017; 6:10.1002/adhm.201700670. [PMID: 29076281 PMCID: PMC5813692 DOI: 10.1002/adhm.201700670] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/29/2017] [Indexed: 12/25/2022]
Abstract
Repair or regeneration of damaged nerves is still a challenging clinical task in reconstructive surgeries and regenerative medicine. Here, it is demonstrated that periodontal ligament stem cells (PDLSCs) and gingival mesenchymal stem cells (GMSCs) isolated from adult human periodontal and gingival tissues assume neuronal phenotype in vitro and in vivo via a subcutaneous transplantation model in nude mice. PDLSCs and GMSCs are encapsulated in a 3D scaffold based on alginate and hyaluronic acid hydrogels capable of sustained release of human nerve growth factor (NGF). The elasticity of the hydrogels affects the proliferation and differentiation of encapsulated MSCs within scaffolds. Moreover, it is observed that PDLSCs and GMSCs are stained positive for βIII-tubulin, while exhibiting high levels of gene expression related to neurogenic differentiation (βIII-tubulin and glial fibrillary acidic protein) via quantitative polymerase chain reaction (qPCR). Western blot analysis shows the importance of elasticity of the matrix and the presence of NGF in the neurogenic differentiation of encapsulated MSCs. In vivo, immunofluorescence staining for neurogenic specific protein markers confirms islands of dense positively stained structures inside transplanted hydrogels. As far as it is known, this study is the first demonstration of the application of PDLSCs and GMSCs as promising cell therapy candidates for nerve regeneration.
Collapse
Affiliation(s)
- Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Ivana M Diniz
- Faculdade de Odontologia da UFMG, Departamento de Odontologia Restauradora, Av. Antonio Carlos, 6627, Belo Horizonte, MG, 31270-910, Brazil
| | - Chider Chen
- School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, PA, 19104, USA
| | - Patricia Sarrion
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, NE 68508, Lincoln
| | - Benjamin M Wu
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
222
|
Xiao D, Yan H, Wang Q, Lv X, Zhang M, Zhao Y, Zhou Z, Xu J, Sun Q, Sun K, Li W, Lu M. Trilayer Three-Dimensional Hydrogel Composite Scaffold Containing Encapsulated Adipose-Derived Stem Cells Promotes Bladder Reconstruction via SDF-1α/CXCR4 Pathway. ACS APPLIED MATERIALS & INTERFACES 2017; 9:38230-38241. [PMID: 29022693 DOI: 10.1021/acsami.7b10630] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Bladder acellular matrix graft-alginate dialdehyde-gelatin hydrogel-silk mesh (BAMG-HS) encapsulated with adipose-derived stem cells (ASCs) was evaluated in a rat model of augmentation cystoplasty, including BAMG-HS-ASCs (n = 18, subgroup n = 6 for 2, 4, and 12 weeks), acellular BAMG-HS (n = 6 for 12 weeks) and cystotomy control (n = 6 for 12 weeks) groups. Equipped with good cytocompatibility and superior mechanical properties (elastic modulus: 5.33 ± 0.96 MPa, maximum load: 28.90 ± 0.69 N), BAMG-HS acted a trilayer "sandwich" scaffold with minimal interference in systemic homeostasis. ASCs in BAMG-HS promoted morphological and histological bladder restoration by accelerating scaffold degradation (p < 0.05), ameliorating fibrosis (p < 0.05) and inflammation (p < 0.01). Additionally, ASCs facilitated the recovery of bladder function by enhancing smooth muscle regeneration (p < 0.05), innervation (p < 0.01) and angiogenesis (p < 0.001). Except for a small number of endothelium-differentiated ASCs, the pro-angiogenic effects of ASCs were mainly related to ERK1/2 phosphorylation in the downstream of SDF-1α/CXCR4 pathway.
Collapse
Affiliation(s)
- Dongdong Xiao
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| | - Hao Yan
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| | - Qiong Wang
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou 510120, China
| | - Xiangguo Lv
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| | - Ming Zhang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| | - Yang Zhao
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| | - Zhe Zhou
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| | - Jiping Xu
- Department of Urology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200011, China
| | - Qian Sun
- The State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Kang Sun
- The State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Wei Li
- The State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Mujun Lu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai 200001, China
| |
Collapse
|
223
|
Anderson EM, Silva EA, Hao Y, Martinick KD, Lewin SA, Stafford AG, Doherty EG, Wang L, Doherty EJ, Grossman PM, Mooney DJ. VEGF and IGF Delivered from Alginate Hydrogels Promote Stable Perfusion Recovery in Ischemic Hind Limbs of Aged Mice and Young Rabbits. J Vasc Res 2017; 54:288-298. [PMID: 28930755 PMCID: PMC5642984 DOI: 10.1159/000479869] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
Biomaterial-based delivery of angiogenic growth factors restores perfusion more effectively than bolus delivery methods in rodent models of peripheral vascular disease, but the same success has not yet been demonstrated in clinically relevant studies of aged or large animals. These studies explore, in clinically relevant models, a therapeutic angiogenesis strategy for the treatment of peripheral vascular disease that overcomes the challenges encountered in previous clinical trials. Alginate hydrogels providing sustained release of vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF) were injected into ischemic hind limbs in middle-aged and old mice, and also in young rabbits, as a test of the scalability of this local growth factor treatment. Spontaneous perfusion recovery diminished with increasing age, and only the combination of VEGF and IGF delivery from gels significantly rescued perfusion in middle-aged (13 months) and old (20 months) mice. In rabbits, the delivery of VEGF alone or in combination with IGF from alginate hydrogels, at a dose 2 orders of magnitude lower than the typical doses used in past rabbit studies, enhanced perfusion recovery when given immediately after surgery, or as a treatment for chronic ischemia. Capillary density measurements and angiographic analysis demonstrated the benefit of gel delivery. These data together suggest that alginate hydrogels providing local delivery of low doses of VEGF and IGF constitute a safe and effective treatment for hind-limb ischemia in clinically relevant animal models, thereby supporting the potential clinical translation of this concept.
Collapse
Affiliation(s)
- Erin M Anderson
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Eduardo A Silva
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Yibai Hao
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Kathleen D Martinick
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Sarah A Lewin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Alexander G Stafford
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Elisabeth G Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Lin Wang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Edward J Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Paul M Grossman
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Dave J Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| |
Collapse
|
224
|
Atelocollagen-based Hydrogels Crosslinked with Oxidised Polysaccharides as Cell Encapsulation Matrix for Engineered Bioactive Stromal Tissue. Tissue Eng Regen Med 2017; 14:539-556. [PMID: 30603508 DOI: 10.1007/s13770-017-0063-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 02/08/2023] Open
Abstract
Tissue stroma is responsible for extracellular matrix (ECM) formation and secretion of factors that coordinate the behaviour of the surrounding cells through the microenvironment created. It's inability to spontaneously regenerate makes it a good candidate for research studies such as testing various tissue engineered products capable of replacing the stroma in order to assure normal tissue regeneration and function. In this study, a bioactive stroma was obtained considering two main components: 1) the artificial ECM formed using atelocollagen-oxidized polysaccharides hydrogels in which the polysaccharide compound (oxidised gellan or pullulan) has the role of crosslinker and 2) encapsulated stromal cells (dermal fibroblasts, ovarian theca-interstitial and granulosa cells). The cell-hosting ability of the hydrogels is demonstrated by a good diffusion of globular proteins (albumin) while the fibrillar morphology proves to be optimal for cell adhesion. These structural properties and cytocompatibility of the components maintain good cell viability and cell encapsulation for more than 12 days. Nevertheless, the results indicate some differences favouring the gellan crosslinked hydrogels. Ovarian stromal cells functionality was maintained as indicated by hormone secretion, confirming cell-cell signalling in encapsulated and co-culture conditions. In vivo implantation shows the regenerative potential of the cell-populated hydrogels as they are integrated into the natural tissue. The possibility of cryopreserving the hydrogel-cell system, while maintaining both cell viability and hydrogel structural integrity underlines the potential of these ready-to-use hydrogels as bioactive stroma for multipurpose tissue regeneration.
Collapse
|
225
|
Kerschenmeyer A, Arlov Ø, Malheiro V, Steinwachs M, Rottmar M, Maniura-Weber K, Palazzolo G, Zenobi-Wong M. Anti-oxidant and immune-modulatory properties of sulfated alginate derivatives on human chondrocytes and macrophages. Biomater Sci 2017; 5:1756-1765. [PMID: 28643827 DOI: 10.1039/c7bm00341b] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Degeneration of articular cartilage represents one of the most common causes of pain and disability in our aging society. Current treatments only address the symptoms of joint disease, but not their underlying causes which include oxidative stress and inflammation in cartilage and surrounding tissues. Sulfated biopolymers that mimic aspects of the native extracellular environment of cartilage are recently gaining interest as a means to slow the inflammatory events responsible for tissue degeneration. Here we show that the natural polysaccharide alginate and particularly its sulfated derivatives have potent anti-oxidant, anti-inflammatory and anti-immunogenic properties in vitro. We found that these polymers exert a free radical scavenging activity in a sulfation-dependent manner. In particular, the sulfation degree of substitution of alginate directly correlated with its ability to scavenge superoxide radicals and to chelate metal ions. We also studied the effect of sulfated alginate on the ability of IL-1β to stimulate inflammatory genes in human chondrocytes and found decreased expression of the pro-inflammatory markers IL-6 and CXCL8, which inversely correlated with the sulfation degree. Moreover, in studies testing the ability of the alginates to modulate macrophage polarization, we found that they decreased both the gene expression and synthesis of the proinflammatory cytokine TNF-α in human THP-1 macrophages with M1-like phenotype in a sulfation-dependent manner. To conclude, sulfated alginates effectively protect against oxidative stress and inflammation in vitro and are a promising biomaterial to be explored for treatment of osteoarthritis.
Collapse
Affiliation(s)
- Anne Kerschenmeyer
- Otto-Stern-Weg 7, Cartilage Engineering+Regeneration, ETH Zurich, 8093 Zurich, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Oliver S, Jofri A, Thomas DS, Vittorio O, Kavallaris M, Boyer C. Tuneable catechin functionalisation of carbohydrate polymers. Carbohydr Polym 2017; 169:480-494. [DOI: 10.1016/j.carbpol.2017.04.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/10/2017] [Accepted: 04/14/2017] [Indexed: 10/19/2022]
|
227
|
Kyle S, Jessop ZM, Al-Sabah A, Whitaker IS. 'Printability' of Candidate Biomaterials for Extrusion Based 3D Printing: State-of-the-Art. Adv Healthc Mater 2017; 6. [PMID: 28558161 DOI: 10.1002/adhm.201700264] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/02/2017] [Indexed: 12/24/2022]
Abstract
Regenerative medicine has been highlighted as one of the UK's 8 'Great Technologies' with the potential to revolutionize patient care in the 21st Century. Over the last decade, the concept of '3D bioprinting' has emerged, which allows the precise deposition of cell laden bioinks with the aim of engineering complex, functional tissues. For 3D printing to be used clinically, there is the need to produce advanced functional biomaterials, a new generation of bioinks with suitable cell culture and high shape/print fidelity, to match or exceed the physical, chemical and biological properties of human tissue. With the rapid increase in knowledge associated with biomaterials, cell-scaffold interactions and the ability to biofunctionalize/decorate bioinks with cell recognition sequences, it is important to keep in mind the 'printability' of these novel materials. In this illustrated review, we define and refine the concept of 'printability' and review seminal and contemporary studies to highlight the current 'state of play' in the field with a focus on bioink composition and concentration, manipulation of nozzle parameters and rheological properties.
Collapse
Affiliation(s)
- Stuart Kyle
- Reconstructive Surgery & Regenerative Medicine Group (ReconRegen); Institute of Life Sciences; Swansea University Medical School; Swansea SA2 8PP UK
- The Welsh Centre for Burns and Plastic Surgery; Morriston Hospital; Swansea SA6 6NL UK
| | - Zita M. Jessop
- Reconstructive Surgery & Regenerative Medicine Group (ReconRegen); Institute of Life Sciences; Swansea University Medical School; Swansea SA2 8PP UK
- The Welsh Centre for Burns and Plastic Surgery; Morriston Hospital; Swansea SA6 6NL UK
| | - Ayesha Al-Sabah
- Reconstructive Surgery & Regenerative Medicine Group (ReconRegen); Institute of Life Sciences; Swansea University Medical School; Swansea SA2 8PP UK
| | - Iain S. Whitaker
- Reconstructive Surgery & Regenerative Medicine Group (ReconRegen); Institute of Life Sciences; Swansea University Medical School; Swansea SA2 8PP UK
- The Welsh Centre for Burns and Plastic Surgery; Morriston Hospital; Swansea SA6 6NL UK
| |
Collapse
|
228
|
Fenn SL, Charron PN, Oldinski RA. Anticancer Therapeutic Alginate-Based Tissue Sealants for Lung Repair. ACS APPLIED MATERIALS & INTERFACES 2017; 9:23409-23419. [PMID: 28648052 PMCID: PMC5546308 DOI: 10.1021/acsami.7b04932] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Injury to the connective tissue that lines the lung, the pleura, or the lung itself can occur from many causes including trauma or surgery, as well as lung diseases or cancers. To address current limitations for patching lung injuries, to stop air or fluid leaks, an adherent hydrogel sealant patch system was developed, based on methacrylated alginate (AMA) and AMA dialdehyde (AMA-DA) blends, which is capable of sealing damaged tissues and sustaining physiological pressures. Methacrylation of alginate hydroxyl groups rendered the polysaccharide capable of photo-cross-linking when mixed with an eosin Y-based photoinitiator system and exposed to visible green light. Oxidation of alginate yields functional aldehyde groups capable of imine bond formation with proteins found in many tissues. The alginate-based patch system was rigorously tested on a custom burst pressure testing device. Blending of nonoxidized material with oxidized (aldehyde modified) alginates yielded patches with improved burst pressure performance and decreased delamination as compared with pure AMA. Human mesothelial cell (MeT-5A) viability and cytotoxicity were retained when cultured with the hydrogel patches. The release and bioactivity of doxorubicin-encapsulated submicrospheres enabled the fabrication of drug-eluting adhesive patches and were effective in decreasing human lung cancer cell (A549) viability.
Collapse
Affiliation(s)
- Spencer L. Fenn
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155
- Bioengineering Program, College of Engineering and Mathematical Sciences, and Larner College of Medicine, University of Vermont, Burlington, VT, 05405
| | - Patrick N. Charron
- Department of Mechanical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, 05405
| | - Rachael A. Oldinski
- Bioengineering Program, College of Engineering and Mathematical Sciences, and Larner College of Medicine, University of Vermont, Burlington, VT, 05405
- Department of Mechanical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, 05405
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, 05405
- Department of Orthopaedics and Rehabilitation, Larner College of Medicine, University of Vermont, Burlington, VT, 05405
| |
Collapse
|
229
|
Alginate hydrogels allow for bioactive and sustained release of VEGF-C and VEGF-D for lymphangiogenic therapeutic applications. PLoS One 2017; 12:e0181484. [PMID: 28723974 PMCID: PMC5517064 DOI: 10.1371/journal.pone.0181484] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/30/2017] [Indexed: 11/19/2022] Open
Abstract
Lymphatic dysfunction is associated with the progression of many cardiovascular disorders due to their role in maintaining tissue fluid homeostasis. Promoting new lymphatic vessels (lymphangiogenesis) is a promising strategy to reverse these cardiovascular disorders via restoring lymphatic function. Vascular endothelial growth factor (VEGF) members VEGF-C and VEGF-D are both potent candidates for stimulating lymphangiogenesis, though maintaining spatial and temporal control of these factors represents a challenge to developing efficient therapeutic lymphangiogenic applications. Injectable alginate hydrogels have been useful for the controlled delivery of many angiogenic factors, including VEGF-A, to stimulate new blood vasculature. However, the utility of these tunable hydrogels for delivering lymphangiogenic factors has never been closely examined. Thus, the objective of this study was to utilize ionically cross-linked alginate hydrogels to deliver VEGF-C and VEGF-D for potential lymphangiogenic applications. We demonstrated that lymphatic endothelial cells (LECs) are sensitive to temporal presentation of VEGF-C and VEGF-D but with different responses between the factors. The greatest LEC mitogenic and sprouting response was observed for constant concentrations of VEGF-C and a high initial concentration that gradually decreased over time for VEGF-D. Additionally, alginate hydrogels provided sustained release of radiolabeled VEGF-C and VEGF-D. Finally, VEGF-C and VEGF-D released from these hydrogels promoted a similar number of LEC sprouts as exogenously added growth factors and new vasculature in vivo via a chick chorioallantoic membrane (CAM) assay. Overall, these findings demonstrate that alginate hydrogels can provide sustained and bioactive release of VEGF-C and VEGF-D which could have applications for therapeutic lymphangiogenesis.
Collapse
|
230
|
Yang J, Zhang YS, Yue K, Khademhosseini A. Cell-laden hydrogels for osteochondral and cartilage tissue engineering. Acta Biomater 2017; 57:1-25. [PMID: 28088667 PMCID: PMC5545789 DOI: 10.1016/j.actbio.2017.01.036] [Citation(s) in RCA: 432] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 12/21/2016] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
Abstract
Despite tremendous advances in the field of regenerative medicine, it still remains challenging to repair the osteochondral interface and full-thickness articular cartilage defects. This inefficiency largely originates from the lack of appropriate tissue-engineered artificial matrices that can replace the damaged regions and promote tissue regeneration. Hydrogels are emerging as a promising class of biomaterials for both soft and hard tissue regeneration. Many critical properties of hydrogels, such as mechanical stiffness, elasticity, water content, bioactivity, and degradation, can be rationally designed and conveniently tuned by proper selection of the material and chemistry. Particularly, advances in the development of cell-laden hydrogels have opened up new possibilities for cell therapy. In this article, we describe the problems encountered in this field and review recent progress in designing cell-hydrogel hybrid constructs for promoting the reestablishment of osteochondral/cartilage tissues. Our focus centers on the effects of hydrogel type, cell type, and growth factor delivery on achieving efficient chondrogenesis and osteogenesis. We give our perspective on developing next-generation matrices with improved physical and biological properties for osteochondral/cartilage tissue engineering. We also highlight recent advances in biomanufacturing technologies (e.g. molding, bioprinting, and assembly) for fabrication of hydrogel-based osteochondral and cartilage constructs with complex compositions and microarchitectures to mimic their native counterparts. STATEMENT OF SIGNIFICANCE Despite tremendous advances in the field of regenerative medicine, it still remains challenging to repair the osteochondral interface and full-thickness articular cartilage defects. This inefficiency largely originates from the lack of appropriate tissue-engineered biomaterials that replace the damaged regions and promote tissue regeneration. Cell-laden hydrogel systems have emerged as a promising tissue-engineering platform to address this issue. In this article, we describe the fundamental problems encountered in this field and review recent progress in designing cell-hydrogel constructs for promoting the reestablishment of osteochondral/cartilage tissues. Our focus centers on the effects of hydrogel composition, cell type, and growth factor delivery on achieving efficient chondrogenesis and osteogenesis. We give our perspective on developing next-generation hydrogel/inorganic particle/stem cell hybrid composites with improved physical and biological properties for osteochondral/cartilage tissue engineering. We also highlight recent advances in biomanufacturing and bioengineering technologies (e.g. 3D bioprinting) for fabrication of hydrogel-based osteochondral and cartilage constructs.
Collapse
Affiliation(s)
- Jingzhou Yang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Guangzhou Women and Children's Medical Center, Sun Yat-sen University, Guangzhou 510623, Guangdong, People's Republic of China
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kan Yue
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| |
Collapse
|
231
|
Owonubi S, Agwuncha S, Mukwevho E, Aderibigbe B, Sadiku E, Biotidara O, Varaprasad K. Application of Hydrogel Biocomposites for Multiple Drug Delivery. HANDBOOK OF COMPOSITES FROM RENEWABLE MATERIALS 2017:139-165. [DOI: 10.1002/9781119441632.ch110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
232
|
Diekjürgen D, Grainger DW. Polysaccharide matrices used in 3D in vitro cell culture systems. Biomaterials 2017; 141:96-115. [PMID: 28672214 DOI: 10.1016/j.biomaterials.2017.06.020] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 12/22/2022]
Abstract
Polysaccharides comprise a diverse class of polymeric materials with a history of proven biocompatibility and continual use as biomaterials. Recent focus on new matrices appropriate for three-dimensional (3D) cell culture offers new opportunities to apply polysaccharides as extracellular matrix mimics. However, chemical and structural bases for specific cell-polysaccharide interactions essential for their utility as 3-D cell matrices are not well defined. This review describes how these naturally sourced biomaterials satisfy several key properties for current 3D cell culture needs and can also be synthetically modified or blended with additional components to tailor their cell engagement properties. Beyond their benign interactions with many cell types in cultures, their economical and high quality sourcing, optical clarity for ex situ analytical interrogation and in situ gelation represent important properties of these polymers for 3D cell culture applications. Continued diversification of their versatile glycan chemistry, new bio-synthetic sourcing strategies and elucidation of new cell-specific properties are attractive to expand the polysaccharide polymer utility for cell culture needs. Many 3D cell culture priorities are addressed with the portfolio of polysaccharide materials available and under development. This review provides a critical analysis of their properties, capabilities and challenges in 3D cell culture applications.
Collapse
Affiliation(s)
- Dorina Diekjürgen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112-5820, USA.
| |
Collapse
|
233
|
Zhang Z, Chai W, Xiong R, Zhou L, Huang Y. Printing-induced cell injury evaluation during laser printing of 3T3 mouse fibroblasts. Biofabrication 2017. [DOI: 10.1088/1758-5090/aa6ed9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
234
|
Zhang Z, Chai W, Xiong R, Zhou L, Huang Y. Printing-induced cell injury evaluation during laser printing of 3T3 mouse fibroblasts. Biofabrication 2017. [DOI: 10.1088/1758-5090/aa6ed9/.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
235
|
Zhang Z, Chai W, Xiong R, Zhou L, Huang Y. Printing-induced cell injury evaluation during laser printing of 3T3 mouse fibroblasts. Biofabrication 2017. [PMID: 28631624 DOI: 10.1088/1758-5090/aa6ed9/] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Three-dimensional bioprinting has emerged as a promising solution for the freeform fabrication of living cellular constructs, which can be used for tissue/organ transplantation and tissue models. During bioprinting, some living cells are unavoidably injured and may become necrotic or apoptotic cells. This study aims to investigate the printing-induced cell injury and evaluates injury types of post-printing cells using the annexin V/7-aminoactinomycin D and FAM-DEVD-FMK/propidium iodide assays during laser printing of NIH 3T3 mouse fibroblasts. As observed, the percentage of post-printing early apoptotic mouse fibroblasts increases with the incubation time, indicating that post-printing apoptotic mouse fibroblasts have different initiation lag times of apoptosis due to different levels of mechanical stress exerted during laser printing. Post-printing necrotic mouse fibroblasts can be detected immediately after printing, while post-printing early apoptotic mouse fibroblasts need time to develop into a late apoptotic stage. The minimum time needed for post-printing early apoptotic mouse fibroblasts to complete their apoptosis pathway and transition into late apoptotic mouse fibroblasts is from 4 h to 5 h post-printing. The resulting knowledge of the evolution of different apoptotic post-printing mouse fibroblasts will help better design future experiments to quantitatively determine, model, and mitigate the post-printing cell injury based on molecular signal pathway modeling.
Collapse
Affiliation(s)
- Zhengyi Zhang
- School of Naval Architecture and Ocean Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China. Dept. of Mechanical and Aerospace Engineering, Univ. of Florida, Gainesville, FL 32611, United States of America
| | | | | | | | | |
Collapse
|
236
|
De Silva RT, Mantilaka MMMGPG, Goh KL, Ratnayake SP, Amaratunga GAJ, de Silva KMN. Magnesium Oxide Nanoparticles Reinforced Electrospun Alginate-Based Nanofibrous Scaffolds with Improved Physical Properties. Int J Biomater 2017; 2017:1391298. [PMID: 28694826 PMCID: PMC5485316 DOI: 10.1155/2017/1391298] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 01/15/2023] Open
Abstract
Mechanically robust alginate-based nanofibrous scaffolds were successfully fabricated by electrospinning method to mimic the natural extracellular matrix structure which benefits development and regeneration of tissues. Alginate-based nanofibres were electrospun from an alginate/poly(vinyl alcohol) (PVA) polyelectrolyte complex. SEM images revealed the spinnability of the complex composite nanofibrous scaffolds, showing randomly oriented, ultrafine, and virtually defects-free alginate-based/MgO nanofibrous scaffolds. Here, it is shown that an alginate/PVA complex scaffold, blended with near-spherical MgO nanoparticles (⌀ 45 nm) at a predetermined concentration (10% (w/w)), is electrospinnable to produce a complex composite nanofibrous scaffold with enhanced mechanical stability. For the comparison purpose, chemically cross-linked electrospun alginate-based scaffolds were also fabricated. Tensile test to rupture revealed the significant differences in the tensile strength and elastic modulus among the alginate scaffolds, alginate/MgO scaffolds, and cross-linked alginate scaffolds (P < 0.05). In contrast to cross-linked alginate scaffolds, alginate/MgO scaffolds yielded the highest tensile strength and elastic modulus while preserving the interfibre porosity of the scaffolds. According to the thermogravimetric analysis, MgO reinforced alginate nanofibrous scaffolds exhibited improved thermal stability. These novel alginate-based/MgO scaffolds are economical and versatile and may be further optimised for use as extracellular matrix substitutes for repair and regeneration of tissues.
Collapse
Affiliation(s)
- R. T. De Silva
- Nanotechnology and Science Park, Sri Lanka Institute of Nanotechnology (SLINTEC), Pitipana, Homagama, Sri Lanka
| | - M. M. M. G. P. G. Mantilaka
- Nanotechnology and Science Park, Sri Lanka Institute of Nanotechnology (SLINTEC), Pitipana, Homagama, Sri Lanka
| | - K. L. Goh
- School of Mechanical and Systems Engineering, Newcastle University, Newcastle Upon Tyne, UK
| | - S. P. Ratnayake
- Nanotechnology and Science Park, Sri Lanka Institute of Nanotechnology (SLINTEC), Pitipana, Homagama, Sri Lanka
| | - G. A. J. Amaratunga
- Nanotechnology and Science Park, Sri Lanka Institute of Nanotechnology (SLINTEC), Pitipana, Homagama, Sri Lanka
- Electrical Engineering Division, Department of Engineering, University of Cambridge, 9 J. J. Thomson Avenue, Cambridge CB3 0FA, UK
| | - K. M. Nalin de Silva
- Nanotechnology and Science Park, Sri Lanka Institute of Nanotechnology (SLINTEC), Pitipana, Homagama, Sri Lanka
- Department of Chemistry, University of Colombo, Colombo 3, Sri Lanka
| |
Collapse
|
237
|
Zhang Z, Xu C, Xiong R, Chrisey DB, Huang Y. Effects of living cells on the bioink printability during laser printing. BIOMICROFLUIDICS 2017; 11:034120. [PMID: 28670353 PMCID: PMC5472480 DOI: 10.1063/1.4985652] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/31/2017] [Indexed: 05/24/2023]
Abstract
Laser-induced forward transfer has been a promising orifice-free bioprinting technique for the direct writing of three-dimensional cellular constructs from cell-laden bioinks. In order to optimize the printing performance, the effects of living cells on the bioink printability must be carefully investigated in terms of the ability to generate well-defined jets during the jet/droplet formation process as well as well-defined printed droplets on a receiving substrate during the jet/droplet deposition process. In this study, a time-resolved imaging approach has been implemented to study the jet/droplet formation and deposition processes when printing cell-free and cell-laden bioinks under different laser fluences. It is found that the jetting behavior changes from no material transferring to well-defined jetting with or without an initial bulgy shape to jetting with a bulgy shape/pluming/splashing as the laser fluence increases. Under desirable well-defined jetting, two impingement-based deposition and printing types are identified: droplet-impingement printing and jet-impingement printing with multiple breakups. Compared with cell-free bioink printing, the transfer threshold of the cell-laden bioink is higher while the jet velocity, jet breakup length, and printed droplet size are lower, shorter, and smaller, respectively. The addition of living cells transforms the printing type from jet-impingement printing with multiple breakups to droplet-impingement printing. During the printing of cell-laden bioinks, two non-ideal jetting behaviors, a non-straight jet with a non-straight trajectory and a straight jet with a non-straight trajectory, are identified mainly due to the local nonuniformity and nonhomogeneity of cell-laden bioinks.
Collapse
Affiliation(s)
| | - Changxue Xu
- Department of Industrial, Manufacturing, and Systems Engineering, Texas Tech University, Lubbock, Texas 79409, USA
| | - Ruitong Xiong
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Douglas B Chrisey
- Department of Physics and Engineering Physics, Tulane University, New Orleans, Louisiana 70118, USA
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
| |
Collapse
|
238
|
Barron C, He JQ. Alginate-based microcapsules generated with the coaxial electrospray method for clinical application. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1245-1255. [PMID: 28391767 DOI: 10.1080/09205063.2017.1318030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Alginate-based microencapsulation of cells has made a significant impact on the fields of regenerative medicine and tissue engineering mainly because of its ability to provide immunoisolation for the encapsulated material. This characteristic has allowed for the successful transplantation of non-autologous cells in several clinical trials for life threatening conditions, such as diabetes, myocardial infarction, and neurodegenerative disorders. Methods for alginate hydrogel microencapsulation have been well developed for various types of cells and can generate microcapsules of different diameters, degradation time, and composition. It appears the most prominent and successful method in clinical applications is the coaxial electrospray method, which can be used to generate both homogenous and non-homogeneous microcapsules with uniform size on the order of 100 μm. The present review aims to discuss why alginate hydrogel is an ideal biomaterial for the encapsulation of cells, how alginate-based microcapsules are generated, and methods of modifying the microcapsules for specific clinical treatments. This review will also discuss clinical applications that have utilized alginate-based microencapsulation in the treatment of diabetes, ischemic heart disease, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Catherine Barron
- a Department of Biomedical Sciences & Pathobiology , College of Veterinary Medicine, Virginia Polytechnic Institute and State University , Blacksburg , VA , USA
| | - Jia-Qiang He
- a Department of Biomedical Sciences & Pathobiology , College of Veterinary Medicine, Virginia Polytechnic Institute and State University , Blacksburg , VA , USA
| |
Collapse
|
239
|
Ansari S, Seagroves JT, Chen C, Shah K, Aghaloo T, Wu BM, Bencharit S, Moshaverinia A. Dental and orofacial mesenchymal stem cells in craniofacial regeneration: The prosthodontist's point of view. J Prosthet Dent 2017; 118:455-461. [PMID: 28385446 DOI: 10.1016/j.prosdent.2016.11.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022]
Abstract
Of the available regenerative treatment options, craniofacial tissue regeneration using mesenchymal stem cells (MSCs) shows promise. The ability of stem cells to produce multiple specialized cell types along with their extensive distribution in many adult tissues have made them an attractive target for applications in tissue engineering. MSCs reside in a wide spectrum of postnatal tissue types and have been successfully isolated from orofacial tissues. These dental- or orofacial-derived MSCs possess self-renewal and multilineage differentiation capacities. The craniofacial system is composed of complex hard and soft tissues derived from sophisticated processes starting with embryonic development. Because of the complexity of the craniofacial tissues, the application of stem cells presents challenges in terms of the size, shape, and form of the engineered structures, the specialized final developed cells, and the modulation of timely blood supply while limiting inflammatory and immunological responses. The cell delivery vehicle has an important role in the in vivo performance of stem cells and could dictate the success of the regenerative therapy. Among the available hydrogel biomaterials for cell encapsulation, alginate-based hydrogels have shown promising results in biomedical applications. Alginate scaffolds encapsulating MSCs can provide a suitable microenvironment for cell viability and differentiation for tissue regeneration applications. This review aims to summarize current applications of dental-derived stem cell therapy and highlight the use of alginate-based hydrogels for applications in craniofacial tissue engineering.
Collapse
Affiliation(s)
- Sahar Ansari
- Lecturer, Division of Oral Biology, School of Dentistry, University of California, Los Angeles, Calif
| | - Jackson T Seagroves
- Student, Department of Dental Research, School of Dentistry, University of North Carolina, Chapel Hill, NC
| | - Chider Chen
- Postdoctoral research fellow, Department of Anatomy and Cell Biology, School of Dental Medicine University of Pennsylvania, Philadelphia, Pa
| | - Kumar Shah
- Associate Professor and Program Director, Graduate Program in Prosthodontics, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Calif
| | - Tara Aghaloo
- Professor, Division of Advanced Prosthodontics and Director, Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, Calif
| | - Benjamin M Wu
- Professor and Chair, Division of Advanced Prosthodontics and Director, Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, Calif
| | - Sompop Bencharit
- Associate Professor and Director, Digital Dentistry Technologies, Department of General Practice and Department of Oral & Maxillofacial Surgery, School of Dentistry, and Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA
| | - Alireza Moshaverinia
- Assistant Professor, Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Calif.
| |
Collapse
|
240
|
Song Y, Zhang C, Wang P, Wang L, Bao C, Weir MD, Reynolds MA, Ren K, Zhao L, Xu HHK. Engineering bone regeneration with novel cell-laden hydrogel microfiber-injectable calcium phosphate scaffold. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 75:895-905. [PMID: 28415545 DOI: 10.1016/j.msec.2017.02.158] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/26/2017] [Accepted: 02/27/2017] [Indexed: 02/05/2023]
Abstract
Cell-based tissue engineering is promising to create living functional tissues for bone regeneration. The implanted cells should be evenly distributed in the scaffold, be fast-released to the defect and maintain high viability in order to actively participate in the regenerative process. Herein, we report an injectable calcium phosphate cement (CPC) scaffold containing cell-encapsulating hydrogel microfibers with desirable degradability that could deliver cells in a timely manner and maintain cell viability. Microfibers were synthesized using partially-oxidized alginate with various concentrations (0-0.8%) of fibrinogen to optimize the degradation rate of the alginate-fibrin microfibers (Alg-Fb MF). A fibrin concentration of 0.4% in Alg-Fb MF resulted in the greatest enhancement of cell migration, release and proliferation. Interestingly, a significant amount of cell-cell contact along the long-axis of the microfibers was established in Alg-0.4%Fb MF as early as day 2. The injectable tissue engineered construct for bone reconstruct was fabricated by mixing the fast-degradable Alg-0.4%Fb MF with CPC paste at 1:1 volume ratio. In vitro study showed that cells re-collected from the construct maintained good viability and osteogenic potentials. In vivo study demonstrated that the hBMSC-encapsulated CPC-MF tissue engineered construct displayed a robust capacity for bone regeneration. At 12weeks after implantation, osseous bridge in the rat mandibular defect was observed in CPC-MF-hBMSCs group with a new bone area fraction of (42.1±7.8) % in the defects, which was >3-fold that of the control group. The novel tissue-engineered construct presents an excellent prospect for a wide range of dental, craniofacial and orthopedic applications.
Collapse
Affiliation(s)
- Yang Song
- Department of Prosthodontics, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Chi Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ping Wang
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA.
| | - Lin Wang
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130011, China
| | - Chunyun Bao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Michael D Weir
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Mark A Reynolds
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Liang Zhao
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Hockin H K Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
241
|
Mistry P, Aied A, Alexander M, Shakesheff K, Bennett A, Yang J. Bioprinting Using Mechanically Robust Core-Shell Cell-Laden Hydrogel Strands. Macromol Biosci 2017; 17. [PMID: 28160431 DOI: 10.1002/mabi.201600472] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/23/2016] [Indexed: 11/06/2022]
Abstract
The strand material in extrusion-based bioprinting determines the microenvironments of the embedded cells and the initial mechanical properties of the constructs. One unmet challenge is the combination of optimal biological and mechanical properties in bioprinted constructs. Here, a novel bioprinting method that utilizes core-shell cell-laden strands with a mechanically robust shell and an extracellular matrix-like core has been developed. Cells encapsulated in the strands demonstrate high cell viability and tissue-like functions during cultivation. This process of bioprinting using core-shell strands with optimal biochemical and biomechanical properties represents a new strategy for fabricating functional human tissues and organs.
Collapse
Affiliation(s)
- Pritesh Mistry
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ahmed Aied
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Morgan Alexander
- Division of Surface Analysis and Biophysics, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Kevin Shakesheff
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Andrew Bennett
- FRAME Laboratory, School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Jing Yang
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
242
|
Dalheim MØ, Ulset AST, Jenssen IB, Christensen BE. Degradation kinetics of peptide-coupled alginates prepared via the periodate oxidation reductive amination route. Carbohydr Polym 2017; 157:1844-1852. [DOI: 10.1016/j.carbpol.2016.11.068] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/18/2016] [Accepted: 11/23/2016] [Indexed: 11/26/2022]
|
243
|
Chen H, Xing X, Tan H, Jia Y, Zhou T, Chen Y, Ling Z, Hu X. Covalently antibacterial alginate-chitosan hydrogel dressing integrated gelatin microspheres containing tetracycline hydrochloride for wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:287-295. [DOI: 10.1016/j.msec.2016.08.086] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 08/12/2016] [Accepted: 08/31/2016] [Indexed: 02/07/2023]
|
244
|
|
245
|
Challenges for Cartilage Regeneration. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2017. [DOI: 10.1007/978-3-662-53574-5_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
246
|
Gilbert WJR, Johnson SJ, Tsau JS, Liang JT, Scurto AM. Enzymatic degradation of polyacrylamide in aqueous solution with peroxidase and H2O2. J Appl Polym Sci 2016. [DOI: 10.1002/app.44560] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- William J. R. Gilbert
- University of Kansas, Department of Chemical & Petroleum Engineering; Lawrence Kansas 66045
| | - Stephen J. Johnson
- University of Kansas, Department of Chemical & Petroleum Engineering; Lawrence Kansas 66045
| | - Jyun-Syung Tsau
- Tertiary Oil Recovery Program; University of Kansas; Lawrence Kansas 66045
| | - Jenn-Tai Liang
- University of Kansas, Department of Chemical & Petroleum Engineering; Lawrence Kansas 66045
- Tertiary Oil Recovery Program; University of Kansas; Lawrence Kansas 66045
| | - Aaron M. Scurto
- University of Kansas, Department of Chemical & Petroleum Engineering; Lawrence Kansas 66045
| |
Collapse
|
247
|
Wang L, Wang P, Weir MD, Reynolds MA, Zhao L, Xu HHK. Hydrogel fibers encapsulating human stem cells in an injectable calcium phosphate scaffold for bone tissue engineering. ACTA ACUST UNITED AC 2016; 11:065008. [PMID: 27811389 DOI: 10.1088/1748-6041/11/6/065008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs), human embryonic stem cells (hESCs) and human umbilical cord mesenchymal stem cells (hUCMSCs) are exciting cell sources for use in regenerative medicine. There have been no reports on long hydrogel fibers encapsulating stem cells inside an injectable calcium phosphate cement (CPC) scaffold for bone tissue engineering. The objectives of this study were: (1) to develop a novel injectable CPC construct containing hydrogel fibers encapsulating cells for bone engineering, and (2) to investigate and compare cell viability, proliferation and osteogenic differentiation of hiPSC-MSCs, hESC-MSCs and hUCMSCs in injectable CPC. The pastes encapsulating the stem cells were fully injectable under a small injection force, and the injection did not harm the cells, compared with non-injected cells (p > 0.1). The mechanical properties of the stem cell-CPC construct were much better than those of previous injectable polymers and hydrogels for cell delivery. The hiPSC-MSCs, hESC-MSCs and hUCMSCs in hydrogel fibers in CPC had excellent proliferation and osteogenic differentiation. All three cell types yielded high alkaline phosphatase, runt-related transcription factor, collagen I and osteocalcin expression (mean ± SD; n = 6). Cell-synthesized minerals increased substantially with time (p < 0.05), with no significant difference among the three types of cells (p > 0.1). Mineralization by hiPSC-MSCs, hESC-MSCs and hUCMSCs in CPC at 14 d was 13-fold that at 1 d. In conclusion, all three types of cells (hiPSC-MSCs, hESC-MSCs and hUCMSCs) in a CPC scaffold showed high potential for bone tissue engineering, and the novel injectable CPC construct with cell-encapsulating hydrogel fibers is promising for enhancing bone regeneration in dental, craniofacial and orthopedic applications.
Collapse
Affiliation(s)
- Lin Wang
- VIP Integrated Department, Stomatological Hospital of Jilin University, Changchun, Jilin 130011, People's Republic of China. Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
248
|
Chen Q, Chen D, Wu J, Lin JM. Flexible control of cellular encapsulation, permeability, and release in a droplet-templated bifunctional copolymer scaffold. BIOMICROFLUIDICS 2016; 10:064115. [PMID: 27990217 PMCID: PMC5148761 DOI: 10.1063/1.4972107] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/30/2016] [Indexed: 05/27/2023]
Abstract
Designing cell-compatible, bio-degradable, and stimuli-responsive hydrogels is very important for biomedical applications in cellular delivery and micro-scale tissue engineering. Here, we report achieving flexible control of cellular microencapsulation, permeability, and release by rationally designing a diblock copolymer, alginate-conjugated poly(N-isopropylacrylamide) (Alg-co-PNiPAM). We use the microfluidic technique to fabricate the bifunctional copolymers into thousands of mono-disperse droplet-templated hydrogel microparticles for controlled encapsulation and triggered release of mammalian cells. In particular, the grafting PNiPAM groups in the synthetic cell-laden microgels produce lots of nano-aggregates into hydrogel networks at elevated temperature, thereafter enhancing the permeability of microparticle scaffolds. Importantly, the hydrogel scaffolds are readily fabricated via on-chip quick gelation by triggered release of Ca2+ from the Ca-EDTA complex; it is also quite exciting that very mild release of microencapsulated cells is achieved via controlled degradation of hydrogel scaffolds through a simple strategy of competitive affinity of Ca2+ from the Ca-Alginate complex. This finding suggests that we are able to control cellular encapsulation and release through ion-induced gelation and degradation of the hydrogel scaffolds. Subsequently, we demonstrate a high viability of microencapsulated cells in the microgel scaffolds.
Collapse
Affiliation(s)
- Qiushui Chen
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University , Beijing, China
| | - Dong Chen
- Institute of Process Equipment, College of Chemical and Biological Engineering, Zhejiang University , Hangzhou, China
| | - Jing Wu
- School of Science, China University of Geosciences (Beijing) , Beijing, China
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University , Beijing, China
| |
Collapse
|
249
|
Jia J, Coyle RC, Richards DJ, Berry CL, Barrs RW, Biggs J, James Chou C, Trusk TC, Mei Y. Development of peptide-functionalized synthetic hydrogel microarrays for stem cell and tissue engineering applications. Acta Biomater 2016; 45:110-120. [PMID: 27612960 PMCID: PMC5146757 DOI: 10.1016/j.actbio.2016.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 10/21/2022]
Abstract
Synthetic polymer microarray technology holds remarkable promise to rapidly identify suitable biomaterials for stem cell and tissue engineering applications. However, most of previous microarrayed synthetic polymers do not possess biological ligands (e.g., peptides) to directly engage cell surface receptors. Here, we report the development of peptide-functionalized hydrogel microarrays based on light-assisted copolymerization of poly(ethylene glycol) diacrylates (PEGDA) and methacrylated-peptides. Using solid-phase peptide/organic synthesis, we developed an efficient route to synthesize methacrylated-peptides. In parallel, we identified PEG hydrogels that effectively inhibit non-specific cell adhesion by using PEGDA-700 (M. W.=700) as a monomer. The combined use of these chemistries enables the development of a powerful platform to prepare peptide-functionalized PEG hydrogel microarrays. Additionally, we identified a linker composed of 4 glycines to ensure sufficient exposure of the peptide moieties from hydrogel surfaces. Further, we used this system to directly compare cell adhesion abilities of several related RGD peptides: RGD, RGDS, RGDSG and RGDSP. Finally, we combined the peptide-functionalized hydrogel technology with bioinformatics to construct a library composed of 12 different RGD peptides, including 6 unexplored RGD peptides, to develop culture substrates for hiPSC-derived cardiomyocytes (hiPSC-CMs), a cell type known for poor adhesion to synthetic substrates. 2 out of 6 unexplored RGD peptides showed substantial activities to support hiPSC-CMs. Among them, PMQKMRGDVFSP from laminin β4 subunit was found to support the highest adhesion and sarcomere formation of hiPSC-CMs. With bioinformatics, the peptide-functionalized hydrogel microarrays accelerate the discovery of novel biological ligands to develop biomaterials for stem cell and tissue engineering applications. STATEMENT OF SIGNIFICANCE In this manuscript, we described the development of a robust approach to prepare peptide-functionalized synthetic hydrogel microarrays. Combined with bioinformatics, this technology enables us to rapidly identify novel biological ligands for the development of the next generation of functional biomaterials for stem cell and tissue engineering applications.
Collapse
Affiliation(s)
- Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert C Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | | | - Ryan Walker Barrs
- College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Joshua Biggs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - C James Chou
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Thomas C Trusk
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
250
|
Zehnder T, Freund T, Demir M, Detsch R, Boccaccini AR. Fabrication of Cell-Loaded Two-Phase 3D Constructs for Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2016; 9:E887. [PMID: 28774008 PMCID: PMC5457208 DOI: 10.3390/ma9110887] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/14/2016] [Accepted: 10/17/2016] [Indexed: 12/23/2022]
Abstract
Hydrogel optimisation for biofabrication considering shape stability/mechanical properties and cell response is challenging. One approach to tackle this issue is to combine different additive manufacturing techniques, e.g., hot-melt extruded thermoplastics together with bioplotted cell loaded hydrogels in a sequential plotting process. This method enables the fabrication of 3D constructs mechanically supported by the thermoplastic structure and biologically functionalised by the hydrogel phase. In this study, polycaprolactone (PCL) and polyethylene glycol (PEG) blend (PCL-PEG) together with alginate dialdehyde gelatine hydrogel (ADA-GEL) loaded with stromal cell line (ST2) were investigated. PCL-PEG blends were evaluated concerning plotting properties to fabricate 3D scaffolds, namely miscibility, wetting behaviour and in terms of cell response. Scaffolds were characterised considering pore size, porosity, strut width, degradation behaviour and mechanical stability. Blends showed improved hydrophilicity and cell response with PEG blending increasing the degradation and decreasing the mechanical properties of the scaffolds. Hybrid constructs with PCL-PEG blend and ADA-GEL were fabricated. Cell viability, distribution, morphology and interaction of cells with the support structure were analysed. Increased degradation of the thermoplastic support structure and proliferation of the cells not only in the hydrogel, but also on the thermoplastic phase, indicates the potential of this novel material combination for biofabricating 3D tissue engineering scaffolds.
Collapse
Affiliation(s)
- Tobias Zehnder
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany.
| | - Tim Freund
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany.
| | - Merve Demir
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany.
| | - Rainer Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany.
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany.
| |
Collapse
|