201
|
Tunable bioactivity and mechanics of collagen-based tissue engineering constructs: A comparison of EDC-NHS, genipin and TG2 crosslinkers. Biomaterials 2020; 254:120109. [PMID: 32480093 PMCID: PMC7298615 DOI: 10.1016/j.biomaterials.2020.120109] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 02/05/2023]
Abstract
Due to its ubiquity and versatility in the human body, collagen is an ideal base material for tissue-engineering constructs. Chemical crosslinking treatments allow precise control of the biochemical and mechanical properties through macromolecular modifications to the structure of collagen. In this work, three key facets regarding the collagen crosslinking process are explored. Firstly, a comparison is drawn between the carbodiimide-succinimide (EDC-NHS) system and two emerging crosslinkers utilising alternate chemistries: genipin and tissue transglutaminase (TG2). By characterising the chemical changes upon treatment, the effect of EDC-NHS, genipin and TG2 crosslinking mechanisms on the chemical structure of collagen, and thus the mechanical properties conferred to the substrate is explored. Secondly, the relative importance of mechanical and biochemical cues on cellular phenomena are investigated, including cell viability, integrin-specific attachment, spreading and proliferation. Here, we observe that for human dermal fibroblasts, long-term, stable proliferation is preconditioned by the availability of suitable binding sites, irrespective of the substrate modulus post-crosslinking. Finally, as seen in the graphical abstract we show that by choosing the appropriate crosslinker chemistries, a materials selection map can be drawn for collagen films, encompassing both a range of tensile modulus and fibroblast proliferation which can be modified independently. Thus, in addition to a range of parameters that can be modified in collagen constructs, we demonstrate a route to obtaining tunable bioactivity and mechanics in collagen constructs is uncovered, that is exclusively driven by the crosslinking process.
Collapse
|
202
|
Santra M, Sharma M, Katoch D, Jain S, Saikia UN, Dogra MR, Luthra-Guptasarma M. Induction of posterior vitreous detachment (PVD) by non-enzymatic reagents targeting vitreous collagen liquefaction as well as vitreoretinal adhesion. Sci Rep 2020; 10:8250. [PMID: 32427865 PMCID: PMC7237681 DOI: 10.1038/s41598-020-64931-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/26/2020] [Indexed: 11/20/2022] Open
Abstract
Induction of posterior vitreous detachment (PVD) by pharmacologic vitreolysis has been largely attempted through the use of enzymatic reagents. Ocriplasmin has been the only FDA-approved clinical reagent so far. Several adverse effects of ocriplasmin have emerged, however, and the search for alternative PVD-inducing reagents continues. Since i) collagen forms an important structural component of the vitreous, and ii) strong vitreo-retinal adhesions exist between the cortical vitreous and the internal limiting membrane (ILM) of the retina, an effective PVD-inducing reagent would require both, vitreous liquefaction, and concurrent dehiscence of vitreoretinal adhesion, without being toxic to retinal cells. We designed a combination of two reagents to achieve these two objectives; a triple helix-destabilizing collagen binding domain (CBD), and a fusion of RGD (integrin-binding) tripeptide with CBD (RCBD) to facilitate separation of posterior cortical vitreous from retinal surface. Based on in vitro, ex-vivo, and in vivo experiments, we show that a combination of CBD and RCBD displays potential for safe pharmacologic vitreolysis. Our findings assume significance in light of the fact that synthetic RGD-containing peptides have already been used for inhibition of tumor cell invasion. Proteins such as variants of collagen binding domains could have extended therapeutic uses in the future.
Collapse
Affiliation(s)
- Mithun Santra
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Maryada Sharma
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.,Department of Otolaryngology and Head & Neck surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Deeksha Katoch
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Sahil Jain
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Uma Nahar Saikia
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Mangat R Dogra
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Manni Luthra-Guptasarma
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|
203
|
Yan X, Zhang K, Yang Y, Deng D, Lyu C, Xu H, Liu W, Du Y. Dispersible and Dissolvable Porous Microcarrier Tablets Enable Efficient Large-Scale Human Mesenchymal Stem Cell Expansion. Tissue Eng Part C Methods 2020; 26:263-275. [PMID: 32268824 DOI: 10.1089/ten.tec.2020.0039] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) have wide applications in regenerative medicine but their clinical translation is largely hindered by limited production capacity of current cell expansion regime. This study utilizes a novel dispersible and dissolvable porous microcarrier tablet, 3D TableTrix™, in stirred bioreactor to demonstrate a scalable expansion protocol for industrial manufacturing of hMSCs. The 3D TableTrix is a ready-to-use tablet that disperses into 10s of 1000s porous microcarriers upon contact with culture media, eliminating the need to prepare microcarriers before cell seeding, hence simplifying operation process. We demonstrate over 500 times expansion of adipose-derived hMSCs using serum-free culture medium in 11 days with bead-to-bead transfer for a partial scale-up from laboratory-scale spinner flasks to a 1-L bioreactor system. A final yield of 1.05 ± 0.11 × 109 hMSCs was achieved, and yield of over 3 × 109 with an overall expansion factor of 1530 could theoretically be realized with full scale-up. Cells were harvested by dissolving microcarriers with 98.6% ± 0.1% recovery rate. Cells retained their immunophenotypic characteristics, trilineage differentiation potential, and genome stability with low indications of senescence phenotype. This study illuminates the potential of industrializing clinical-grade hMSC production using 3D TableTrix microcarrier tablets and stirred tank bioreactors. Impact statement The 3D TableTrix™ is a newly available microcarrier ingeniously designed as dispersible and dissolvable porous microcarrier tablets for human mesenchymal stem cell (hMSC) expansion. This eliminates the need of tedious preparation work usually required for microcarriers and its dissolvable nature allows for high cell recovery rate of 98.6% ± 0.1%. Over 500 times expansion of adipose-derived mesenchymal stem cells in serum-free culture media using a 1-L bioreactor system demonstrates its tremendous potential for industrial production of hMSCs.
Collapse
Affiliation(s)
- Xiaojun Yan
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| | - Kun Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yanping Yang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| | - Dongkai Deng
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| | - Cheng Lyu
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing, China
| | - Huanye Xu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China.,Beijing CytoNiche Biotechnology Co. Ltd., Beijing, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
204
|
Amini N, Paluh JL, Xie Y, Saxena V, Sharfstein ST. Insulin production from hiPSC-derived pancreatic cells in a novel wicking matrix bioreactor. Biotechnol Bioeng 2020; 117:2247-2261. [PMID: 32314809 DOI: 10.1002/bit.27359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/01/2020] [Accepted: 04/18/2020] [Indexed: 12/13/2022]
Abstract
Clinical use of pancreatic β islets for regenerative medicine applications requires mass production of functional cells. Current technologies are insufficient for large-scale production in a cost-efficient manner. Here, we evaluate advantages of a porous cellulose scaffold and demonstrate scale-up to a wicking matrix bioreactor as a platform for culture of human endocrine cells. Scaffold modifications were evaluated in a multiwell platform to find the optimum surface condition for pancreatic cell expansion followed by bioreactor culture to confirm suitability. Preceding scale-up, cell morphology, viability, and proliferation of primary pancreatic cells were evaluated. Two optimal surface modifications were chosen and evaluated further for insulin secretion, cell morphology, and viable cell density for human-induced pluripotent stem cell-derived pancreatic cells at different stages of differentiation. Scale-up was accomplished with uncoated, amine-modified cellulose in a miniature bioreactor, and insulin secretion and cell metabolic profiles were determined for 13 days. We achieved 10-fold cell expansion in the bioreactor along with a significant increase in insulin secretion compared with cultures on tissue culture plastic. Our findings define a new method for expansion of pancreatic cells a on wicking matrix cellulose platform to advance cell therapy biomanufacturing for diabetes.
Collapse
Affiliation(s)
- Nooshin Amini
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York
| | - Janet L Paluh
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York
| | - Yubing Xie
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York
| | | | - Susan T Sharfstein
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York
| |
Collapse
|
205
|
No YJ, Castilho M, Ramaswamy Y, Zreiqat H. Role of Biomaterials and Controlled Architecture on Tendon/Ligament Repair and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904511. [PMID: 31814177 DOI: 10.1002/adma.201904511] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 11/10/2019] [Indexed: 06/10/2023]
Abstract
Engineering synthetic scaffolds to repair and regenerate ruptured native tendon and ligament (T/L) tissues is a significant engineering challenge due to the need to satisfy both the unique biological and biomechanical properties of these tissues. Long-term clinical outcomes of synthetic scaffolds relying solely on high uniaxial tensile strength are poor with high rates of implant rupture and synovitis. Ideal biomaterials for T/L repair and regeneration need to possess the appropriate biological and biomechanical properties necessary for the successful repair and regeneration of ruptured tendon and ligament tissues.
Collapse
Affiliation(s)
- Young Jung No
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Yogambha Ramaswamy
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
206
|
Dosta P, Ferber S, Zhang Y, Wang K, Ros A, Uth N, Levinson Y, Abraham E, Artzi N. Scale-up manufacturing of gelatin-based microcarriers for cell therapy. J Biomed Mater Res B Appl Biomater 2020; 108:2937-2949. [PMID: 32356942 DOI: 10.1002/jbm.b.34624] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/11/2020] [Accepted: 04/13/2020] [Indexed: 12/20/2022]
Abstract
Microcarriers, including crosslinked porous gelatin beads (Cultispher G) are widely used as cell carriers for cell therapy applications. Microcarriers can support a range of adherent cell types in stirred tank bioreactor culture, which is scalable up to several thousands of liters. Cultispher G in particular is advantageous for cell therapy applications because it can be dissolved enzymatically, and thus cells can be harvested without the need to perform a large-scale cell-bead filtration step. This enzymatic dissolution, however, is challenged by the slow degradation of the carriers in the presence of enzymes as new extracellular matrix is being deposited by the proliferating cells. This extended dissolution timelimits the yield of cell recovery while compromising cellular viability. We report herein the development of crosslinked porous gelatin beads that afford rapid, stimuli-triggered dissolution for facile cell removal using human mesenchymal stem cells (hMSC) as a model system. We successfully fabricated redox-sensitive beads (RS beads) and studied their cell growth, dissolution time and cell yield, compared to regular gelatin-based beads (Reg beads). We have shown that RS beads allow for much faster dissolution compared to Reg beads, supporting better hMSC detachment and recovery following 8 days of culture in spinner flasks, or in 3L bioreactors. These newly synthesized RS beads show promise as cellular microcarriers and can be used for scale-up manufacturing of different cell types while providing on-demand degradation for facile cell retrieval.
Collapse
Affiliation(s)
- Pere Dosta
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shiran Ferber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi Zhang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kui Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Albert Ros
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicholas Uth
- Research and Technology, Walkersville, Maryland, USA
| | | | - Eytan Abraham
- Research and Technology, Walkersville, Maryland, USA
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
207
|
Semenycheva LL, Egorikhina MN, Chasova VO, Valetova NB, Kuznetsova YL, Mitin AV. Enzymatic Hydrolysis of Marine Collagen and Fibrinogen Proteins in the Presence of Thrombin. Mar Drugs 2020; 18:E208. [PMID: 32290502 PMCID: PMC7230862 DOI: 10.3390/md18040208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 01/07/2023] Open
Abstract
: Enzymatic hydrolysis of native collagen and fibrinogen was carried out under comparable conditions at room temperature. The molecular weight parameters of proteins before and after hydrolysis by thrombin were monitored by gel-penetrating chromatography (GPC). An analysis of the experiment results shows that the molecular weight parameters of the initial fibrinogen (Fn) and cod collagen (CC) are very similar. High molecular CC decays within the first minute, forming two low molecular fractions. The main part (~80%) falls on the fraction with a value of Mw less than 10 kDa. The initial high molecular fraction of Fn with Mw ~320-340 kDa is not completely hydrolyzed even after three days of control. The presence of low molecular fractions with Mw ~17 and Mw ~10 kDa in the solution slightly increases within an hour and noticeably increases for three days. The destruction of macromolecules of high molecular collagen to hydrolysis products appears almost completely within the first minute mainly to the polymer with Mw ~10 kDa, and enzymatic hydrolysis of fibrinogen proceeds slower than that of collagen, but also mainly to the polymer with Mw ~10 kDa. Comparative photos of the surfaces of native collagen, fibrinogen and the scaffold based on them were obtained.
Collapse
Affiliation(s)
- Ludmila L Semenycheva
- Faculty of Chemistry, Lobachevsky State University of Nizhny Novgorod, pr. Gagarina 23, 603950 Nizhny Novgorod, Russia
| | - Marfa N Egorikhina
- Federal State Budgetary Educational Institution of Higher Education Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky square 10/1, 603950 Nizhny Novgorod, Russia
| | - Victoria O Chasova
- Faculty of Chemistry, Lobachevsky State University of Nizhny Novgorod, pr. Gagarina 23, 603950 Nizhny Novgorod, Russia
| | - Natalya B Valetova
- Faculty of Chemistry, Lobachevsky State University of Nizhny Novgorod, pr. Gagarina 23, 603950 Nizhny Novgorod, Russia
| | - Yulia L Kuznetsova
- Faculty of Chemistry, Lobachevsky State University of Nizhny Novgorod, pr. Gagarina 23, 603950 Nizhny Novgorod, Russia
| | - Alexander V Mitin
- Faculty of Chemistry, Lobachevsky State University of Nizhny Novgorod, pr. Gagarina 23, 603950 Nizhny Novgorod, Russia
| |
Collapse
|
208
|
Rumian Ł, Wolf-Brandstetter C, Rößler S, Reczyńska K, Tiainen H, Haugen HJ, Scharnweber D, Pamuła E. Sodium alendronate loaded poly(l-lactide- co-glycolide) microparticles immobilized on ceramic scaffolds for local treatment of bone defects. Regen Biomater 2020; 7:293-302. [PMID: 32523731 PMCID: PMC7266661 DOI: 10.1093/rb/rbaa012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/21/2020] [Accepted: 03/02/2020] [Indexed: 12/15/2022] Open
Abstract
Bone tissue regeneration in critical-size defects is possible after implantation of a 3D scaffold and can be additionally enhanced once the scaffold is enriched with drugs or other factors supporting bone remodelling and healing. Sodium alendronate (Aln), a widely used anti-osteoporosis drug, exhibits strong inhibitory effect on bone resorption performed by osteoclasts. Thus, we propose a new approach for the treatment of bone defects in craniofacial region combining biocompatible titanium dioxide scaffolds and poly(l-lactide-co-glycolide) microparticles (MPs) loaded with Aln. The MPs were effectively attached to the surface of the scaffolds’ pore walls by human recombinant collagen. Drug release from the scaffolds was characterized by initial burst (24 ± 6% of the drug released within first 24 h) followed by a sustained release phase (on average 5 µg of Aln released per day from Day 3 to Day 18). In vitro tests evidenced that Aln at concentrations of 5 and 2.5 µg/ml was not cytotoxic for MG-63 osteoblast-like cells (viability between 81 ± 6% and 98 ± 3% of control), but it prevented RANKL-induced formation of osteoclast-like cells from macrophages derived from peripheral blood mononuclear cells, as shown by reduced fusion capability and decreased tartrate-resistant acid phosphatase 5b activity (56 ± 5% reduction in comparison to control after 8 days of culture). Results show that it is feasible to design the scaffolds providing required doses of Aln inhibiting osteoclastogenesis, reducing osteoclast activity, but not affecting osteoblast functions, which may be beneficial in the treatment of critical-size bone tissue defects.
Collapse
Affiliation(s)
- Łucja Rumian
- Faculty of Materials Science and Ceramics, Department of Biomaterials and Composites, AGH University of Science and Technology, Al. A. Mickiewicza 30, Krakow 30-059, Poland
| | - Cornelia Wolf-Brandstetter
- Technische Universität Dresden, Institute of Materials Science, Max Bergmann Center of Biomaterials, Budapester Str. 27, Dresden 01-069, Germany
| | - Sina Rößler
- Technische Universität Dresden, Institute of Materials Science, Max Bergmann Center of Biomaterials, Budapester Str. 27, Dresden 01-069, Germany
| | - Katarzyna Reczyńska
- Faculty of Materials Science and Ceramics, Department of Biomaterials and Composites, AGH University of Science and Technology, Al. A. Mickiewicza 30, Krakow 30-059, Poland
| | - Hanna Tiainen
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, Geitmyrsveien 71, Blindern, P.O. Box 1109, Oslo NO-0317, Norway
| | - Håvard J Haugen
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, Geitmyrsveien 71, Blindern, P.O. Box 1109, Oslo NO-0317, Norway
| | - Dieter Scharnweber
- Technische Universität Dresden, Institute of Materials Science, Max Bergmann Center of Biomaterials, Budapester Str. 27, Dresden 01-069, Germany
| | - Elżbieta Pamuła
- Faculty of Materials Science and Ceramics, Department of Biomaterials and Composites, AGH University of Science and Technology, Al. A. Mickiewicza 30, Krakow 30-059, Poland
| |
Collapse
|
209
|
Suraiya AB, Hun ML, Truong VX, Forsythe JS, Chidgey AP. Gelatin-Based 3D Microgels for In Vitro T Lineage Cell Generation. ACS Biomater Sci Eng 2020; 6:2198-2208. [PMID: 33455336 DOI: 10.1021/acsbiomaterials.9b01610] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
T cells are predominantly produced by the thymus and play a significant role in maintaining our adaptive immune system. Physiological involution of the thymus occurs gradually with age, compromising naive T cell output, which can have severe clinical complications. Also, T cells are utilized as therapeutic agents in cancer immunotherapies. Therefore, there is an increasing need for strategies aimed at generating naive T cells. The majority of in vitro T cell generation studies are performed in two-dimensional (2D) cultures, which ignore the physiological thymic microenvironment and are not scalable; therefore, we applied a new three-dimensional (3D) approach. Here, we use a gelatin-based 3D microgel system for T lineage induction by co-culturing OP9-DL4 cells and mouse fetal-liver-derived hematopoietic stem cells (HSCs). Flow cytometric analysis revealed that microgel co-cultures supported T lineage induction similar to 2D cultures while providing a 3D environment. We also encapsulated mouse embryonic thymic epithelial cells (TECs) within the microgels to provide a defined 3D culture platform. The microgel system supported TEC maintenance and retained their phenotype. Together, these data show that our microgel system has the capacity for TEC maintenance and induction of in vitro T lineage differentiation with potential for scalability.
Collapse
Affiliation(s)
- Anisha B Suraiya
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia.,Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Michael L Hun
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Vinh X Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Ann P Chidgey
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| |
Collapse
|
210
|
Electrospinning Live Cells Using Gelatin and Pullulan. Bioengineering (Basel) 2020; 7:bioengineering7010021. [PMID: 32098366 PMCID: PMC7148470 DOI: 10.3390/bioengineering7010021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/30/2022] Open
Abstract
Electrospinning is a scaffold production method that utilizes electric force to draw a polymer solution into nanometer-sized fibers. By optimizing the polymer and electrospinning parameters, a scaffold is created with the desired thickness, alignment, and pore size. Traditionally, cells and biological constitutes are implanted into the matrix of the three-dimensional scaffold following electrospinning. Our design simultaneously introduces cells into the scaffold during the electrospinning process at 8 kV. In this study, we achieved 90% viability of adipose tissue-derived stem cells through electrospinning.
Collapse
|
211
|
Sumathy B, Nair PD. Keratinocytes-hair follicle bulge stem cells-fibroblasts co-cultures on a tri-layer skin equivalent derived from gelatin/PEG methacrylate nanofibers. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:869-894. [DOI: 10.1080/09205063.2020.1725861] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Babitha Sumathy
- Division of Tissue Engineering and Regeneration Technologies, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Prabha D Nair
- Division of Tissue Engineering and Regeneration Technologies, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| |
Collapse
|
212
|
Abdollahi Boraei SB, Nourmohammadi J, Bakhshandeh B, Dehghan MM, Gholami H, Calle Hernández D, Gonzalez Z, Ferrari B. Enhanced osteogenesis of gelatin-halloysite nanocomposite scaffold mediated by loading strontium ranelate. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1725754] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - Jhamak Nourmohammadi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Hossein Gholami
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Daniel Calle Hernández
- Gregorio Marañón Health Research Institute, Madrid, Spain
- National Center for Cardiovascular Research Carlos III (CNIC), Madrid, Spain
| | | | | |
Collapse
|
213
|
Gallo N, Lunetti P, Bettini S, Barca A, Madaghiele M, Valli L, Capobianco L, Sannino A, Salvatore L. Assessment of physico-chemical and biological properties of sericin-collagen substrates for PNS regeneration. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1725755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Nunzia Gallo
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Paola Lunetti
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Simona Bettini
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Amilcare Barca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marta Madaghiele
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Ludovico Valli
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Alessandro Sannino
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Luca Salvatore
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| |
Collapse
|
214
|
Elshishiny F, Mamdouh W. Fabrication of Nanofibrous/Xerogel Layer-by-Layer Biocomposite Scaffolds for Skin Tissue Regeneration: In Vitro Study. ACS OMEGA 2020; 5:2133-2147. [PMID: 32064374 PMCID: PMC7016933 DOI: 10.1021/acsomega.9b02832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/20/2020] [Indexed: 06/10/2023]
Abstract
Skin burn wounds are a crucial issue that could reduce life quality. Although numerous effective skin products have invaded the biomedical market, most of them still demonstrate some limitations regarding their porosity, swelling and degradation behaviors, antibacterial properties, and cytotoxicity. Thus, the aim of this study is to fabricate novel trilayered asymmetric porous scaffolds that can mimic the natural skin layers. In particular, the fabricated scaffold constitutes an upper electrospun chitosan-poly(vinyl alcohol) layer and a lower xerogel layer, which is made of effective skin extracellular matrix components. Both layers are fixed together using fibrin glue as a middle layer. The results of this study revealed promising scaffold swelling capability suitable for absorbing wound exudates, followed by a constant degradable weight over time, which is appropriate for a burn wound environment. Scanning electron microscopy images revealed an average pore diameter in the range of 138.39-170.18 nm for the cross-linked electrospun mats and an average pore size of 2.29-30.62 μm for the fabricated xerogel layers. This further provided an optimum environment for fibroblast migration and proliferation. The electrospun nanofibrous layer was examined for its antibacterial properties and showed expressive complete bacterial inhibition against Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli) bacterial strains (log reduction = 3 and 2.70, respectively). Next, mouse embryonic fibroblast cytotoxicity and migration rate were investigated against the developed asymmetrical composite to assess its biocompatibility. Tissue culture experiments demonstrated significant cell proliferation and migration in the presence of the constructed scaffold (P < 0.0001). A complete wound closure was observed in vitro in the presence of the three scaffold asymmetrical layers against the mouse embryonic fibroblast. The results of this study proved superior biological characteristics of the innovative asymmetrical composite that could further replace the burned or damaged skin layers with promising potential for clinical applications.
Collapse
Affiliation(s)
| | - Wael Mamdouh
- E-mail: . Tel: +202
2615 2555. Fax: +202 2797 4951
| |
Collapse
|
215
|
Bello AB, Kim D, Kim D, Park H, Lee SH. Engineering and Functionalization of Gelatin Biomaterials: From Cell Culture to Medical Applications. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:164-180. [PMID: 31910095 DOI: 10.1089/ten.teb.2019.0256] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Health care and medicine were revolutionized in recent years by the development of biomaterials, such as stents, implants, personalized drug delivery systems, engineered grafts, cell sheets, and other transplantable materials. These materials not only support the growth of cells before transplantation but also serve as replacements for damaged tissues in vivo. Among the various biomaterials available, those made from natural biological sources such as extracellular proteins (collagen, fibronectin, laminin) have shown significant benefits, and thus are widely used. However, routine biomaterial-based research requires copious quantities of proteins and the use of pure and intact extracellular proteins could be highly cost ineffective. Gelatin is a molecular derivative of collagen obtained through the irreversible denaturation of collagen proteins. Gelatin shares a very close molecular structure and function with collagen and thus is often used in cell and tissue culture to replace collagen for biomaterial purposes. Recent technological advancements such as additive manufacturing, rapid prototyping, and three-dimensional printing, in general, have resulted in great strides toward the generation of functional gelatin-based materials for medical purposes. In this review, the structural and molecular similarities of gelatin to other extracellular matrix proteins are compared and analyzed. Current strategies for gelatin crosslinking and production are described and recent applications of gelatin-based biomaterials in cell culture and tissue regeneration are discussed. Finally, recent improvements in gelatin-based biomaterials for medical applications and future directions are elaborated. Impact statement In this study, we described gelatin's biochemical properties and compared its advantages and drawbacks over other extracellular matrix proteins and polymers used for biomaterial application. We also described how gelatin can be used with other polymers in creating gelatin composite materials that have enhanced mechanical properties, increased biocompatibility, and boosted bioactivity, maximizing its benefits for biomedical purposes. The article is relevant, as it discussed not only the chemistry of gelatin, but also listed the current techniques in gelatin/biomaterial manufacturing and described the most recent trends in gelatin-based biomaterials for biomedical applications.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.,Department of Biomedical Science, Dongguk University, Gyeonggi, Republic of Korea
| | - Deogil Kim
- Department of Biomedical Science, CHA University, Seongnam-Si, Republic of Korea
| | - Dohyun Kim
- Department of Biomedical Science, Dongguk University, Gyeonggi, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, Dongguk University, Gyeonggi, Republic of Korea
| |
Collapse
|
216
|
Jumelle C, Gholizadeh S, Annabi N, Dana R. Advances and limitations of drug delivery systems formulated as eye drops. J Control Release 2020; 321:1-22. [PMID: 32027938 DOI: 10.1016/j.jconrel.2020.01.057] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
Topical instillation of eye drops remains the most common and easiest route of ocular drug administration, representing the treatment of choice for many ocular diseases. Nevertheless, low ocular bioavailability of topically applied drug molecules can considerably limit their efficacy. Over the last several decades, numerous drug delivery systems (DDS) have been developed in order to improve drug bioavailability on the ocular surfaces. This review systematically covers the most recent advances of DDS applicable by topical instillation, that have shown better performance in in vivo models compared to standard eye drop formulations. These delivery systems are based on in situ forming gels, nanoparticles and combinations of both. Most of the DDS have been developed using natural or synthetic polymers. Polymers offer many advantageous properties for designing advanced DDS including biocompatibility, gelation properties and/or mucoadhesiveness. However, despite the high number of studies published over the last decade, there are several limitations for clinical translation of DDS. This review article focuses on the recent advances for the development of ocular drug delivery systems. In addtion, the potential challenges for commercialization of new DDS are presented.
Collapse
Affiliation(s)
- Clotilde Jumelle
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shima Gholizadeh
- Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, USA
| | - Nasim Annabi
- Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, USA; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, CA, USA.
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
217
|
Sghayyar HN, Lim SS, Ahmed I, Lai JY, Cheong XY, Chong ZW, Lim AFX, Loh HS. Fish biowaste gelatin coated phosphate-glass fibres for wound-healing application. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2019.109386] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
218
|
Carthew J, Donderwinkel I, Shrestha S, Truong V, Forsythe J, Frith J. In situ miRNA delivery from a hydrogel promotes osteogenesis of encapsulated mesenchymal stromal cells. Acta Biomater 2020; 101:249-261. [PMID: 31722255 DOI: 10.1016/j.actbio.2019.11.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/26/2019] [Accepted: 11/07/2019] [Indexed: 11/24/2022]
Abstract
Hydrogels are attractive candidates for use in tissue-engineering and the encapsulation and subsequent differentiation of mesenchymal stem/stromal cells (MSCs) is a strategy that holds great promise for the repair and regeneration of bone and cartilage. However, MSCs are well-known for their sensitivity to mechanical cues, particularly substrate stiffness, and so the inherent softness of hydrogels is poorly matched to the mechanical cues that drive efficient osteogenesis. One approach to overcome this limitation is to harness mechanotransductive signalling pathways and override the signals cells receive from their environment. Previous reports demonstrate that mechanosensitive miRNAs, miR-100-5p and miR-143-3p can enhance MSC osteogenesis, using a complex multi-step procedure to transfect, encapsulate and differentiate the cells. In this study, we develop and characterise a facile system for in situ transfection of MSCs encapsulated within a light-crosslinkable gelatin-PEG hydrogel. Comparing the influence of different transfection agents and hydrogel compositions, we show that particle size, charge, and hydrogel mechanical properties all influence the diffusion of embedded transfection agent complexes. By incorporating both MSCs and transfection agents into the hydrogels we demonstrate successful in situ transfection of encapsulated MSCs. Comparing the efficacy of pre- and in situ transfection of miR-100-5p/miR-143-3p on the osteogenic capacity of hydrogel-encapsulated MSCs, our data demonstrates superior mineralisation and osteogenic gene expression following in situ transfections. Overall, we demonstrate a simple, one-pot system for in situ transfection of miRNAs to enhance MSC osteogenic potential and thus demonstrates significant promise to improve the efficiency of MSC differentiation in hydrogels for bone tissue-engineering applications. STATEMENT OF SIGNIFICANCE: Mesenchymal stromal cells (MSCs) are sensitive to cues from their surrounding microenvironment. Osteogenesis is enhanced in MSCs grown on stiffer substrates, but this is limited when using hydrogels for bone tissue-engineering. Modulating pro-osteogenic genes with mechanosensitive microRNAs (miRNAs) represents a potential tool to overcome this challenge. Here we report a hydrogel platform to deliver miRNAs to encapsulated MSCs. We characterise effects of hydrogel composition and transfection agent type on their mobility and transfection efficiency, demonstrating successful in situ transfection of MSCs and showing that miRNAs can significantly enhance osteogenic mineral deposition and marker gene expression. This system was simpler and more effective than conventional 2D transfection prior to encapsulation and therefore holds promise to improve MSC differentiation in bone tissue-engineering.
Collapse
|
219
|
Azarudeen RS, Hassan MN, Yassin MA, Thirumarimurugan M, Muthukumarasamy N, Velauthapillai D, Mustafa K. 3D printable Polycaprolactone-gelatin blends characterized for in vitro osteogenic potency. REACT FUNCT POLYM 2020. [DOI: 10.1016/j.reactfunctpolym.2019.104445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
220
|
Semenycheva LL, Egorikhina MN, Chasova VO, Valetova NB, Podguzkova MV, Astanina MV, Kuznetsova YL. Enzymatic hydrolysis of collagen by pancreatin and thrombin as a step in the formation of scaffolds. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2738-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
221
|
Fabrication and evaluation of modified poly(ethylene terephthalate) microfibrous scaffolds for hepatocyte growth and functionality maintenance. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 109:110523. [PMID: 32228959 DOI: 10.1016/j.msec.2019.110523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
For hepatocyte culture in vitro, the surface feature of utilized scaffolds exerts a direct impact on cell adhesion, growth and differentiated functionality. Herein, to regulate hepatocyte growth and differentiated functionality, modified microfibrous scaffolds were fabricated by surface grafting monoamine terminated lactobionic lactone (L-NH2) and gelatin onto non-woven poly(ethylene terephthalate) (PET) fibrous substrate (PET-Gal and PET-Gel), respectively. The physicochemical properties of PET scaffolds before and after modification were characterized. Upon 15-day culture, the effects of modified PET scaffolds on growth and differentiated functionality of human induced hepatocytes (hiHeps) were evaluated, compared with that of control without modification. Results demonstrated that both L-NH2 and gelatin modifications improved scaffold properties including hydrophilicity, water uptake ratio, stiffness and roughness, resulting in efficient cell adhesion, ~20-fold cell expansion and enhanced differentiated functionality. After culture for 15 days, PET-Gal cultured cells formed aggregates, displaying better cell viability and significantly higher differentiated functionality regarding albumin secretion, urea synthesis, phases I (cytochrome P450, CYP1A1/2 and CYP3A4) and II (uridine 5'-diphosphate glucuronosyltransferases, UGT) enzyme activity, biliary excretion and detoxification ability (ammonia elimination and bilirubin conjugation), compared with PET and PET-Gel cultured ones. Hence, as a three-dimensional (3D) microfibrous scaffold, PET-Gal promotes hiHeps growth and differentiated functionality maintenance, which is promisingly utilized in bioartificial liver (BAL) bioreactors.
Collapse
|
222
|
Oh SY, Choi DH, Jin YM, Yu Y, Kim HY, Kim G, Park YS, Jo I. Optimization of Microenvironments Inducing Differentiation of Tonsil-Derived Mesenchymal Stem Cells into Endothelial Cell-Like Cells. Tissue Eng Regen Med 2019; 16:631-643. [PMID: 31824825 PMCID: PMC6879685 DOI: 10.1007/s13770-019-00221-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/04/2019] [Accepted: 09/17/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Stem cell engineering is appealing consideration for regenerating damaged endothelial cells (ECs) because stem cells can differentiate into EC-like cells. In this study, we demonstrate that tonsil-derived mesenchymal stem cells (TMSCs) can differentiate into EC-like cells under optimal physiochemical microenvironments. METHODS TMSCs were preconditioned with Dulbecco's Modified Eagle Medium (DMEM) or EC growth medium (EGM) for 4 days and then replating them on Matrigel to observe the formation of a capillary-like network under light microscope. Microarray, quantitative real time polymerase chain reaction, Western blotting and immunofluorescence analyses were used to evaluate the expression of gene and protein of EC-related markers. RESULTS Preconditioning TMSCs in EGM for 4 days and then replating them on Matrigel induced the formation of a capillary-like network in 3 h, but TMSCs preconditioned with DMEM did not form such a network. Genome analyses confirmed that EGM preconditioning significantly affected the expression of genes related to angiogenesis, blood vessel morphogenesis and development, and vascular development. Western blot analyses revealed that EGM preconditioning with gelatin coating induced the expression of endothelial nitric oxide synthase (eNOS), a mature EC-specific marker, as well as phosphorylated Akt at serine 473, a signaling molecule related to eNOS activation. Gelatin-coating during EGM preconditioning further enhanced the stability of the capillary-like network, and also resulted in the network more closely resembled to those observed in human umbilical vein endothelial cells. CONCLUSION This study suggests that under specific conditions, i.e., EGM preconditioning with gelatin coating for 4 days followed by Matrigel, TMSCs could be a source of generating endothelial cells for treating vascular dysfunction.
Collapse
Affiliation(s)
- Se-Young Oh
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Da Hyeon Choi
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Chungdae-ro 1, Seowon-Gu, Cheongju, Chungbuk 28644 Republic of Korea
| | - Yoon Mi Jin
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Yeonsil Yu
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Ha Yeong Kim
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Gyungah Kim
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| | - Yoon Shin Park
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Chungdae-ro 1, Seowon-Gu, Cheongju, Chungbuk 28644 Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
- Ewha Tonsil-derived Mesenchymal Stem Cells Research Center (ETSRC), College of Medicine, Ewha Womans University, 260 Gonghang-daero, Gangseo-gu Seoul, 07804 Republic of Korea
| |
Collapse
|
223
|
Egorikhina MN, Aleynik DY, Rubtsova YP, Levin GY, Charykova IN, Semenycheva LL, Bugrova ML, Zakharychev EA. Hydrogel scaffolds based on blood plasma cryoprecipitate and collagen derived from various sources: Structural, mechanical and biological characteristics. Bioact Mater 2019; 4:334-345. [PMID: 31720490 PMCID: PMC6838346 DOI: 10.1016/j.bioactmat.2019.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/04/2019] [Accepted: 10/13/2019] [Indexed: 01/19/2023] Open
Abstract
At present there is a growing need for tissue engineering products, including the products of scaffold-technologies. Biopolymer hydrogel scaffolds have a number of advantages and are increasingly being used to provide means of cell transfer for therapeutic treatments and for inducing tissue regeneration. This work presents original hydrogel biopolymer scaffolds based on a blood plasma cryoprecipitate and collagen and formed under conditions of enzymatic hydrolysis. Two differently originated collagens were used for the scaffold formation. During this work the structural and mechanical characteristics of the scaffold were studied. It was found that, depending on the origin of collagen, scaffolds possess differences in their structural and mechanical characteristics. Both types of hydrogel scaffolds have good biocompatibility and provide conditions that maintain the three-dimensional growth of adipose tissue stem cells. Hence, scaffolds based on such a blood plasma cryoprecipitate and collagen have good prospects as cell carriers and can be widely used in regenerative medicine.
Collapse
Affiliation(s)
- Marfa N. Egorikhina
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Diana Ya Aleynik
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Yulia P. Rubtsova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Grigory Ya Levin
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Irina N. Charykova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | | | - Marina L. Bugrova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | | |
Collapse
|
224
|
Dai Y, Guo H, Chu L, He Z, Wang M, Zhang S, Shang X. Promoting osteoblasts responses in vitro and improving osteointegration in vivo through bioactive coating of nanosilicon nitride on polyetheretherketone. J Orthop Translat 2019; 24:198-208. [PMID: 33101971 PMCID: PMC7548345 DOI: 10.1016/j.jot.2019.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/20/2019] [Accepted: 10/28/2019] [Indexed: 12/02/2022] Open
Abstract
Objective To enhance the bioactivity of polyetheretherketone (PEEK) while maintain its mechanical strengths. Methods Suspension coating and melt bonding. Results Silicon nitride (Si3N4, SN) coating lead to higher surface roughness, hydrophilicity and protein absorption; SN coating could slowly release Si ion into simulated body fluid (SBF), which caused weak alkaline of micro-environment owing to the slight dissolution of SN; SN coating resulted in the improvements of adhesion, proliferation, differentiation and gene expressions of MC3T3-E1 cells in vitro; SN coating of PEEK with bioactive SN coating (CSNPK) obviously promoted bone regeneration and osseointegration in vivo. Conclusions CSNPK with SN coating as bone implant might be a promising candidate for orthopedic implants. The Translational Potential of this Article The silicon nitride-coated polyetheretherketone (CSNPK) prepared in this article could induce MC3T3-E1 cells adhesion, proliferation and differentiation in vitro; it could also induce bone regeneration in bone defect in vivo, which indicate its good cytocompatibility and biocompatibility. If the raw materials are medical grade, and preparation process as well as production process of this article are further improved, it will have great translational potential.
Collapse
Affiliation(s)
- Yong Dai
- Shandong University, Jinan, 250012, Shandong, China
| | - Han Guo
- Shanghai Institute of Applied Physics, CAS, 2019 Jialuo Road, Shanghai, 201800, China.,Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, CAS, 239 Zhangheng Road, Shanghai, 201204, China
| | - Linyang Chu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zihao He
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Minqi Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shuhong Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xifu Shang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| |
Collapse
|
225
|
Fields MA, Del Priore LV, Adelman RA, Rizzolo LJ. Interactions of the choroid, Bruch's membrane, retinal pigment epithelium, and neurosensory retina collaborate to form the outer blood-retinal-barrier. Prog Retin Eye Res 2019; 76:100803. [PMID: 31704339 DOI: 10.1016/j.preteyeres.2019.100803] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023]
Abstract
The three interacting components of the outer blood-retinal barrier are the retinal pigment epithelium (RPE), choriocapillaris, and Bruch's membrane, the extracellular matrix that lies between them. Although previously reviewed independently, this review integrates these components into a more wholistic view of the barrier and discusses reconstitution models to explore the interactions among them. After updating our understanding of each component's contribution to barrier function, we discuss recent efforts to examine how the components interact. Recent studies demonstrate that claudin-19 regulates multiple aspects of RPE's barrier function and identifies a barrier function whereby mutations of claudin-19 affect retinal development. Co-culture approaches to reconstitute components of the outer blood-retinal barrier are beginning to reveal two-way interactions between the RPE and choriocapillaris. These interactions affect barrier function and the composition of the intervening Bruch's membrane. Normal or disease models of Bruch's membrane, reconstituted with healthy or diseased RPE, demonstrate adverse effects of diseased matrix on RPE metabolism. A stumbling block for reconstitution studies is the substrates typically used to culture cells are inadequate substitutes for Bruch's membrane. Together with human stem cells, the alternative substrates that have been designed offer an opportunity to engineer second-generation culture models of the outer blood-retinal barrier.
Collapse
Affiliation(s)
- Mark A Fields
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Ron A Adelman
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA; Department of Surgery, Yale University School of Medicine, PO Box 208062, New Haven, CT, 06520-8062, USA.
| |
Collapse
|
226
|
Muscle tissue engineering in fibrous gelatin: implications for meat analogs. NPJ Sci Food 2019; 3:20. [PMID: 31646181 PMCID: PMC6803664 DOI: 10.1038/s41538-019-0054-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Bioprocessing applications that derive meat products from animal cell cultures require food-safe culture substrates that support volumetric expansion and maturation of adherent muscle cells. Here we demonstrate scalable production of microfibrous gelatin that supports cultured adherent muscle cells derived from cow and rabbit. As gelatin is a natural component of meat, resulting from collagen denaturation during processing and cooking, our extruded gelatin microfibers recapitulated structural and biochemical features of natural muscle tissues. Using immersion rotary jet spinning, a dry-jet wet-spinning process, we produced gelatin fibers at high rates (~ 100 g/h, dry weight) and, depending on process conditions, we tuned fiber diameters between ~ 1.3 ± 0.1 μm (mean ± SEM) and 8.7 ± 1.4 μm (mean ± SEM), which are comparable to natural collagen fibers. To inhibit fiber degradation during cell culture, we crosslinked them either chemically or by co-spinning gelatin with a microbial crosslinking enzyme. To produce meat analogs, we cultured bovine aortic smooth muscle cells and rabbit skeletal muscle myoblasts in gelatin fiber scaffolds, then used immunohistochemical staining to verify that both cell types attached to gelatin fibers and proliferated in scaffold volumes. Short-length gelatin fibers promoted cell aggregation, whereas long fibers promoted aligned muscle tissue formation. Histology, scanning electron microscopy, and mechanical testing demonstrated that cultured muscle lacked the mature contractile architecture observed in natural muscle but recapitulated some of the structural and mechanical features measured in meat products.
Collapse
|
227
|
Tanase CE, Qutachi O, White LJ, Shakesheff KM, McCaskie AW, Best SM, Cameron RE. Targeted protein delivery: carbodiimide crosslinking influences protein release from microparticles incorporated within collagen scaffolds. Regen Biomater 2019; 6:279-287. [PMID: 31616565 PMCID: PMC6783698 DOI: 10.1093/rb/rbz015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Tissue engineering response may be tailored via controlled, sustained release of active agents from protein-loaded degradable microparticles incorporated directly within three-dimensional (3D) ice-templated collagen scaffolds. However, the effects of covalent crosslinking during scaffold preparation on the availability and release of protein from the incorporated microparticles have not been explored. Here, we load 3D ice-templated collagen scaffolds with controlled additions of poly-(DL-lactide-co-glycolide) microparticles. We probe the effects of subsequent N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride crosslinking on protein release, using microparticles with different internal protein distributions. Fluorescein isothiocyanate labelled bovine serum albumin is used as a model protein drug. The scaffolds display a homogeneous microparticle distribution, and a reduction in pore size and percolation diameter with increased microparticle addition, although these values did not fall below those reported as necessary for cell invasion. The protein distribution within the microparticles, near the surface or more deeply located within the microparticles, was important in determining the release profile and effect of crosslinking, as the surface was affected by the carbodiimide crosslinking reaction applied to the scaffold. Crosslinking of microparticles with a high proportion of protein at the surface caused both a reduction and delay in protein release. Protein located within the bulk of the microparticles, was protected from the crosslinking reaction and no delay in the overall release profile was seen.
Collapse
Affiliation(s)
- Constantin Edi Tanase
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge Centre for Medical Materials, Cambridge, 27, Charles Babbage Road, UK
| | - Omar Qutachi
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, University Park, Nottingham, UK
| | - Lisa J White
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, University Park, Nottingham, UK
| | - Kevin M Shakesheff
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, University Park, Nottingham, UK
| | - Andrew W McCaskie
- Division of Trauma & Orthopaedic Surgery, Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, UK
| | - Serena M Best
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge Centre for Medical Materials, Cambridge, 27, Charles Babbage Road, UK
| | - Ruth E Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge Centre for Medical Materials, Cambridge, 27, Charles Babbage Road, UK
| |
Collapse
|
228
|
Mishra R, Varshney R, Das N, Sircar D, Roy P. Synthesis and characterization of gelatin-PVP polymer composite scaffold for potential application in bone tissue engineering. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.07.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
229
|
Hasturk O, Kaplan DL. Cell armor for protection against environmental stress: Advances, challenges and applications in micro- and nanoencapsulation of mammalian cells. Acta Biomater 2019; 95:3-31. [PMID: 30481608 PMCID: PMC6534491 DOI: 10.1016/j.actbio.2018.11.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
Abstract
Unlike unicellular organisms and plant cells surrounded with a cell wall, naked plasma membranes of mammalian cells make them more susceptible to environmental stresses encountered during in vitro biofabrication and in vivo cell therapy applications. Recent advances in micro- and nanoencapsulation of single mammalian cells provide an effective strategy to isolate cells from their surroundings and protect them against harsh environmental conditions. Microemulsification and droplet-based microfluidics have enabled researchers to encapsulate single cells within a variety of microscale hydrogel materials with a range of biochemical and mechanical properties and functionalities including enhanced cell-matrix interactions or on-demand degradation. In addition to microcapsules, nanocoatings of various organic and inorganic substances on mammalian cells have allowed for the formation of protective shells. A wide range of synthetic and natural polymers, minerals and supramolecular metal-organic complexes have been deposited as nanolayers on the cells via electrostatic interactions, receptor-ligand binding, non-specific interactions, and in situ polymerization/crosslinking. Here, current strategies in encapsulation of single mammalian cells along with challenges and advances are reviewed. Protection of encapsulated stem cells, fibroblasts, red and white blood cells and cancer cells against harsh in vitro and in vivo conditions including anoikis, UV radiation, physical forces, proteolytic enzymes and immune clearance are discussed. STATEMENT OF SIGNIFICANCE: The mechanical fragility of the plasma membrane and susceptibility to extracellular biochemical factors due to the lack of a physical barrier like a tough cell wall or exoskeleton make mammalian cells extra sensitive to harsh environmental conditions. This sensitively, in turn, limits the ex vivo storage, handling and manipulation of mammalian cells, as well as their in vivo applications. Environmental stresses such as exposure to UV, reactive chemicals and mechanical stress during biofabrication processes like 3D bioprinting can often compromise cell viability and function. Micro- and nanoencapsulation of single mammalian cells in protective shells have emerged as promising approaches to isolate cells from their surroundings and enhance resistance against perturbations in conditions during regenerative medicine and tissue engineering applications. In this review, the current state of art of single cell encapsulation strategies and the challenges associated with these technologies are discussed in detail. This is followed by the review of the protection provided by cell armor against a range of harsh in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
230
|
Nair M, Calahorra Y, Kar-Narayan S, Best SM, Cameron RE. Self-assembly of collagen bundles and enhanced piezoelectricity induced by chemical crosslinking. NANOSCALE 2019; 11:15120-15130. [PMID: 31369017 PMCID: PMC7745105 DOI: 10.1039/c9nr04750f] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/15/2019] [Indexed: 05/26/2023]
Abstract
The piezoelectricity of collagen is purported to be linked to many biological processes including bone formation and wound healing. Although the piezoelectricity of tissue-derived collagen has been documented across the length scales, little work has been undertaken to characterise the local electromechanical properties of processed collagen, which is used as a base for tissue-engineering implants. In this work, three chemically distinct treatments used to form structurally and mechanically stable scaffolds-EDC-NHS, genipin and tissue transglutaminase-are investigated for their effect on collagen piezolectricity. Crosslinking with EDC-NHS is noted to produce a distinct self-assembly of the fibres into bundles roughly 300 nm in width regardless of the collagen origin. These fibre bundles also show a localised piezoelectric response, with enhanced vertical piezoelectricity of collagen. Such topographical features are not observed with the other two chemical treatments, although the shear piezoelectric response is significantly enhanced upon crosslinking. These observations are reconciled by a proposed effect of the crosslinking mechanisms on the molecular and nanostructure of collagen. These results highlight the ability to modify the electromechanical properties of collagen using chemical crosslinking methods.
Collapse
Affiliation(s)
- Malavika Nair
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Yonatan Calahorra
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Sohini Kar-Narayan
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Serena M. Best
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Ruth E. Cameron
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| |
Collapse
|
231
|
Osteostimulatory effect of biocomposite scaffold containing phytomolecule diosmin by Integrin/FAK/ERK signaling pathway in mouse mesenchymal stem cells. Sci Rep 2019; 9:11900. [PMID: 31417150 PMCID: PMC6695412 DOI: 10.1038/s41598-019-48429-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/30/2019] [Indexed: 01/01/2023] Open
Abstract
Non-availability of an ideal alternative for autografts in treating critical-size bone defects is a major challenge in orthopedics. Phytocompounds have been proven to enhance osteogenesis via various osteogenic signaling pathways, but its decreased bioavailability and increased renal clearance limit its application. In this study, we designed a biocomposite scaffold comprising gelatin (Gel) and nanohydroxyapatite (nHAp) incorporated with diosmin (DM) and we investigated its bone forming potential in vitro and in vivo. Physiochemical characterization of the scaffold showed that DM had no effect on altering the material characteristics of the scaffold. The addition of DM enhanced the osteoblast differentiation potential of the scaffold in mouse mesenchymal stem cells at both cellular and molecular levels, possibly via the integrin-mediated activation of FAK and ERK signaling components. Using the rat tibial bone defective model, we identified the effect of DM in Gel/nHAp scaffold on enhancing bone formation in vivo. Based on our results, we suggest that Gel/nHAp/DM can be a potential therapeutic agent in scaffold-mediated bone regeneration.
Collapse
|
232
|
Studying nucleic envelope and plasma membrane mechanics of eukaryotic cells using confocal reflectance interferometric microscopy. Nat Commun 2019; 10:3652. [PMID: 31409824 PMCID: PMC6692322 DOI: 10.1038/s41467-019-11645-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Mechanical stress on eukaryotic nucleus has been implicated in a diverse range of diseases including muscular dystrophy and cancer metastasis. Today, there are very few non-perturbative methods to quantify nuclear mechanical properties. Interferometric microscopy, also known as quantitative phase microscopy (QPM), is a powerful tool for studying red blood cell biomechanics. The existing QPM tools, however, have not been utilized to study biomechanics of complex eukaryotic cells either due to lack of depth sectioning, limited phase measurement sensitivity, or both. Here, we present depth-resolved confocal reflectance interferometric microscopy as the next generation QPM to study nuclear and plasma membrane biomechanics. The proposed system features multiple confocal scanning foci, affording 1.5 micron depth-resolution and millisecond frame rate. Furthermore, a near common-path interferometer enables quantifying nanometer-scale membrane fluctuations with better than 200 picometers sensitivity. Our results present accurate quantification of nucleic envelope and plasma membrane fluctuations in embryonic stem cells. Biomechanical studies of eukaryotic cells have been limited due to low sensitivity and axial resolution in interferometric imaging. Here, the authors present depth-resolved confocal reflectance interferometric microscopy with high sensitivity and temporal resolution, which enables quantification of nucleic envelope and plasma membrane fluctuations.
Collapse
|
233
|
Jiang S, Lyu C, Zhao P, Li W, Kong W, Huang C, Genin GM, Du Y. Cryoprotectant enables structural control of porous scaffolds for exploration of cellular mechano-responsiveness in 3D. Nat Commun 2019; 10:3491. [PMID: 31375674 PMCID: PMC6677882 DOI: 10.1038/s41467-019-11397-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/10/2019] [Indexed: 01/09/2023] Open
Abstract
Despite the wide applications, systematic mechanobiological investigation of 3D porous scaffolds has yet to be performed due to the lack of methodologies for decoupling the complex interplay between structural and mechanical properties. Here, we discover the regulatory effect of cryoprotectants on ice crystal growth and use this property to realize separate control of the scaffold pore size and stiffness. Fibroblasts and macrophages are sensitive to both structural and mechanical properties of the gelatin scaffolds, particularly to pore sizes. Interestingly, macrophages within smaller and softer pores exhibit pro-inflammatory phenotype, whereas anti-inflammatory phenotype is induced by larger and stiffer pores. The structure-regulated cellular mechano-responsiveness is attributed to the physical confinement caused by pores or osmotic pressure. Finally, in vivo stimulation of endogenous fibroblasts and macrophages by implanted scaffolds produce mechano-responses similar to the corresponding cells in vitro, indicating that the physical properties of scaffolds can be leveraged to modulate tissue regeneration. Cellular responses to mechanical stimulation have emerged as an important area of research. Here, the authors use cryoprotectant to control the pore size and mechanical properties of porous scaffolds without changing the scaffold composition to allow for the study of cellular mechano-responsiveness in 3D.
Collapse
Affiliation(s)
- Shumeng Jiang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng Lyu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Peng Zhao
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Wenyu Kong
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Chenyu Huang
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Guy M Genin
- Department of Mechanical engineering and Material Science, Washington University at St. Louis, St. Louis, 63130, USA
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
234
|
Ng S, Tan WJ, Pek MMX, Tan MH, Kurisawa M. Mechanically and chemically defined hydrogel matrices for patient-derived colorectal tumor organoid culture. Biomaterials 2019; 219:119400. [PMID: 31398570 DOI: 10.1016/j.biomaterials.2019.119400] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022]
Abstract
Patient-derived tumor organoids offer potentially useful models of cancer tissue physiology. Yet, conventional organoid cultures utilize generic matrices that are difficult to tailor for various unique tumor microenvironments. Here, we employ synthetic, enzymatically crosslinked hydrogels to define mechanical and biochemical properties hypothesized to be relevant for maintaining these organoids. We show that a single extracellular matrix component, gelatin, suffices to support colorectal cancer patient-derived xenograft (CRC-PDX) organoid survival, and that high matrix stiffness synergizes with hypoxia to increase organoid growth and metabolism in a majority of CRC-PDX lines tested. Moreover, we demonstrate that defined gelatin-based hydrogels support CRC-PDX tumor growth in vivo and organoid sensitivity to various CRC therapeutic drugs in vitro in a largely comparable fashion to a conventional reconstituted basement membrane matrix. Based on our findings, we propose that enzymatically crosslinked hydrogels potentially provide a platform for designing mechanically and biochemically defined matrices for various types of patient-derived tumor organoids.
Collapse
Affiliation(s)
- Shengyong Ng
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore, Singapore
| | - Wai Jin Tan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore, Singapore
| | - Michelle Mi Xue Pek
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore, Singapore
| | - Min-Han Tan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore, Singapore
| | - Motoichi Kurisawa
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore, Singapore.
| |
Collapse
|
235
|
Gill E, Willis S, Gerigk M, Cohen P, Zhang D, Li X, Huang YYS. Fabrication of Designable and Suspended Microfibers via Low-Voltage 3D Micropatterning. ACS APPLIED MATERIALS & INTERFACES 2019; 11:19679-19690. [PMID: 31081331 PMCID: PMC6613729 DOI: 10.1021/acsami.9b01258] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/13/2019] [Indexed: 05/02/2023]
Abstract
Building two-dimensional (2D) and three-dimensional (3D) fibrous structures in the micro- and nanoscale will offer exciting prospects for numerous applications spanning from sensors to energy storage and tissue engineering scaffolds. Electrospinning is a well-suited technique for drawing micro- to nanoscale fibers, but current methods of building electrospun fibers in 3D are restrictive in terms of printed height, design of macroscopic fiber networks, and choice of polymer. Here, we combine low-voltage electrospinning and additive manufacturing as a method to pattern layers of suspended mesofibers. Layers of fibers are suspended between 3D-printed supports in situ in multiple fiber layers and designable orientations. We examine the key working parameters to attain a threshold for fiber suspension, use those behavioral observations to establish a "fiber suspension indicator", and demonstrate its utility through design of intricate suspended fiber architectures. Individual fibers produced by this method approach the micrometer/submicrometer scale, while the overall suspended 3D fiber architecture can span over a centimeter in height. We demonstrate an application of suspended fiber architectures in 3D cell culture, utilizing patterned fiber topography to guide the assembly of suspended high-cellular-density structures. The solution-based fiber suspension patterning process we report offers a unique competence in patterning soft polymers, including extracellular matrix-like materials, in a high resolution and aspect ratio. The platform could thus offer new design and manufacturing capabilities of devices and functional products by incorporating functional fibrous elements.
Collapse
Affiliation(s)
- Elisabeth
L. Gill
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| | - Samuel Willis
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Magda Gerigk
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| | - Paul Cohen
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Duo Zhang
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| | - Xia Li
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Yan Yan Shery Huang
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
- The
Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB3 0FF, U.K.
| |
Collapse
|
236
|
Gupta D, Singh AK, Dravid A, Bellare J. Multiscale Porosity in Compressible Cryogenically 3D Printed Gels for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2019; 11:20437-20452. [PMID: 31081613 DOI: 10.1021/acsami.9b05460] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Three-dimensional (3D) printing technology has seen several refinements when introduced in the field of medical devices and regenerative medicines. However, it is still a challenge to 3D print gels for building complex constructs as per the desired shape and size. Here, we present a novel method to 3D print gelatin/carboxymethylchitin/hydroxyapatite composite gel constructs of a complex shape. The objective of this study is to fabricate a bioactive gel scaffold with a controlled hierarchical structure. The hierarchy ranges from 3D outer shape to macroporosity to microporosity and rough surface. The fabrication process developed here uses 3D printing in a local cryogenic atmosphere, followed by lyophilization and cross-linking. The gel instantly freezes after extrusion on the cold plate. The cooling action is not limited to the build plate, but the entire gel scaffold is cooled during the 3D printing process. This enables the construction of a stable self-sustaining large-sized 3D complex geometry. Further, lyophilization introduces bulk microporosity into the scaffolds. The outer shape and macroporosity were controlled with the 3D printer, whereas the microporous structure and desirable rough surface morphology were obtained through lyophilization. With cryogenic 3D printing, up to 90% microporosity could be incorporated into the scaffolds. The microporosity and pore size distribution were controlled by changing the cross-linker and total polymer concentration, which resulted in six times increase in surface open pores of size <20 μm on increasing the cross-linker concentration from 25 to 100 mg/mL. The introduction of bulk microporosity was shown to increase swelling by 1.8 times along with a significant increase in human umbilical cord mesenchymal stem cells and Saos-2 cell attachment (2×), proliferation (2.4×), Saos-2 cell alkaline phosphatase level (2×), and mineralization (3×). The scaffolds are spongy in nature in a wet state, thus making them potential implants for bone cavities with a small opening. The application of these cryogenically 3D printed compressible gel scaffolds with multiscale porosity extends to a small- as well as a large-sized open/partially open patient-specific bone defect.
Collapse
Affiliation(s)
| | - Atul Kumar Singh
- Central Research Facility (CRF) , Indian Institute of Technology Delhi , New Delhi 110016 , India
| | - Ashwin Dravid
- Chemical and Biomolecular Engineering , Johns Hopkins University , 323 E 33rd Street , Baltimore , Maryland 21218 , United States
| | | |
Collapse
|
237
|
Sobhanian P, Khorram M, Hashemi SS, Mohammadi A. Development of nanofibrous collagen-grafted poly (vinyl alcohol)/gelatin/alginate scaffolds as potential skin substitute. Int J Biol Macromol 2019; 130:977-987. [DOI: 10.1016/j.ijbiomac.2019.03.045] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/13/2019] [Accepted: 03/06/2019] [Indexed: 12/31/2022]
|
238
|
Multi-cellular tumor spheroids formation of colorectal cancer cells on Gelatin/PLCL and Collagen/PLCL nanofibrous scaffolds. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
239
|
Smith D, Herman C, Razdan S, Abedin MR, Stoecker WV, Barua S. Microparticles for Suspension Culture of Mammalian Cells. ACS APPLIED BIO MATERIALS 2019; 2:2791-2801. [DOI: 10.1021/acsabm.9b00215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Daniel Smith
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Chase Herman
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Sidharth Razdan
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Muhammad Raisul Abedin
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | | | - Sutapa Barua
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| |
Collapse
|
240
|
Zhang F, Xie Y, Celik H, Akkus O, Bernacki SH, King MW. Engineering small-caliber vascular grafts from collagen filaments and nanofibers with comparable mechanical properties to native vessels. Biofabrication 2019; 11:035020. [PMID: 30943452 DOI: 10.1088/1758-5090/ab15ce] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
At the present time, there is no successful synthetic, off-the-shelf small-caliber vascular graft (<6 mm) for the repair or bypass of the coronary or carotid arteries. This stimulates on-going investigations to fabricate an artificial vascular graft that has both sufficient mechanical properties as well as superior biological performance. Collagen has long been considered as a viable material to encourage cell recruitment, tissue regeneration, and revascularization, but its use has been limited by its inferior mechanical properties. In this study, novel electrochemically aligned collagen filaments were used to engineer a bilayer small-caliber vascular graft, by circular knitting the collagen filaments and electrospinning collagen nanofibers. The collagen prototype grafts showed significantly greater bursting strength under dry and hydrated conditions to that of autografts such as the human internal mammary artery and the saphenous vein (SV). The suture retention strength was sufficient under dry condition, but that under hydrated condition needs to be further improved. The radial dynamic compliance of the collagen grafts was similar to that of the human SV. During in vitro cell culture assays with human umbilical vein endothelial cells, the prototype collagen grafts also encouraged cell adhesion and promoted cell proliferation compared to the synthetic poly(lactic acid) grafts. In conclusion, this study demonstrated the feasibility of the use of novel collagen filaments for fabricating small caliber tissue-engineered vascular grafts that provide both sufficient mechanical properties and superior biological performance.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of Textiles, North Carolina State University, Raleigh, United States of America
| | | | | | | | | | | |
Collapse
|
241
|
Nguyen DT, Dinh VT, Dang LH, Nguyen DN, Giang BL, Nguyen CT, Nguyen TBT, Thu LV, Tran NQ. Dual Interactions of Amphiphilic Gelatin Copolymer and Nanocurcumin Improving the Delivery Efficiency of the Nanogels. Polymers (Basel) 2019; 11:E814. [PMID: 31067644 PMCID: PMC6571557 DOI: 10.3390/polym11050814] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 04/25/2019] [Indexed: 01/14/2023] Open
Abstract
Herein, a new process to manufacture multicore micelles nanoparticles reinforced with co-assembly via hydrophobic interaction and electrostatic interaction under the help of ultrasonication was developed. The precise co-assembly between negative/hydrophobic drug and positive charged amphiphilic copolymer based pluronic platform allows the formation of complex micelles structures as the multicore motif with predefined functions. In this study, curcumin was selected as a drug model while positively charged copolymer was based on a pluronic-conjugated gelatin with different hydrophobicity length of Pluronic F87 and Pluronic F127. Under impact of dual hydrophobic and electrostatic interactions, the nCur-encapsulated core-shell micelles formed ranging from 40 nm to 70 nm and 40-100 nm by transmission electron microscopy (TEM) and Dynamic Light Scattering (DLS), respectively. It is found that the structures emerged depended on the relative lengths of the hydrophobic blocks in pluronic. Regarding g2(τ) behavior from DLS measurement, the nanogels showed a high stability in spherical form. Surprisingly, the release profiles showed a sustainable behavior of Cur from this system for drug delivery approaches. In vitro study exhibited that nCur-encapsulated complex micelles increased inhibitory activity against cancer cells growth with IC50 is 4.02 ± 0.11 mg/L (10.92 ± 0.3 µM) which is higher than of free curcumin at 9.40 ± 0.17 mg/L (25.54 ± 0.18 µM). The results obtained can provide the new method to generate the hierarchical assembly of copolymers with incorporated loading with the same property.
Collapse
Affiliation(s)
- Dinh Trung Nguyen
- Institute of Research and Development, Duy Tan University, Da Nang City 550000, Vietnam.
- Institute of Applied Materials Science, VAST, TL29, ThanhLoc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
| | - Van Thoai Dinh
- Institute of Applied Materials Science, VAST, TL29, ThanhLoc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
- Graduate University of Science and Technology, VAST, TL29, Thanh Loc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
| | - Le Hang Dang
- Institute of Applied Materials Science, VAST, TL29, ThanhLoc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam.
| | - Dang Nam Nguyen
- Institute of Research and Development, Duy Tan University, Da Nang City 550000, Vietnam.
| | - Bach Long Giang
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh, Ward 13, District 4, Ho Chi Minh City 700000, VietNam.
| | - Cong Truc Nguyen
- Institute of Applied Materials Science, VAST, TL29, ThanhLoc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
| | - Thi Bich Tram Nguyen
- Department of Natural Science, Thu Dau Mot University, Thu Dau Mot City 590000, Vietnam.
| | - Le Van Thu
- Institute of Applied Materials Science, VAST, TL29, ThanhLoc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam.
| | - Ngoc Quyen Tran
- Institute of Research and Development, Duy Tan University, Da Nang City 550000, Vietnam.
- Institute of Applied Materials Science, VAST, TL29, ThanhLoc Ward, Dist. 12, Ho Chi Minh City 700000, Vietnam.
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam.
| |
Collapse
|
242
|
Martin CA, Radhakrishnan S, Nagarajan S, Muthukoori S, Dueñas JMM, Gómez Ribelles JL, Lakshmi BS, E A K N, Gómez-Tejedor JA, Reddy MS, Sellathamby S, Rela M, Subbaraya NK. An innovative bioresorbable gelatin based 3D scaffold that maintains the stemness of adipose tissue derived stem cells and the plasticity of differentiated neurons. RSC Adv 2019; 9:14452-14464. [PMID: 35519343 PMCID: PMC9064131 DOI: 10.1039/c8ra09688k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/05/2019] [Indexed: 12/02/2022] Open
Abstract
Neural tissue engineering aims at producing a simulated environment using a matrix that is suitable to grow specialized neurons/glial cells pertaining to CNS/PNS which replace damaged or lost tissues. The primary goal of this study is to design a compatible scaffold that supports the development of neural-lineage cells which aids in neural regeneration. The fabricated, freeze-dried scaffolds consisted of biocompatible, natural and synthetic polymers: gelatin and polyvinyl pyrrolidone. Physiochemical characterization was carried out using Fourier Transform Infrared Spectroscopy (FT-IR) and Scanning Electron Microscopy (SEM) imaging. The 3D construct retains good swelling proficiency and holds the integrated structure that supports cell adhesion and proliferation. The composite of PVP-gelatin is blended in such a way that it matches the mechanical strength of the brain tissue. The cytocompatibility analysis shows that the scaffolds are compatible and permissible for the growth of both stem cells as well as differentiated neurons. A change in the ratios of the scaffold components resulted in varied sizes of pores giving diverse surface morphology, greatly influencing the properties of the neurons. However, there is no change in stem cell properties. Different types of neurons are characterized by the type of gene associated with the neurotransmitter secreted by them. The change in the neuron properties could be attributed to neuroplasticity. The plasticity of the neurons was analyzed using quantitative gene expression studies. It has been observed that the gelatin-rich construct supports the prolonged proliferation of stem cells and multiple neurons along with their plasticity.
Collapse
Affiliation(s)
- Catherine Ann Martin
- Crystal Growth Centre, Anna University Chennai India
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
| | - Subathra Radhakrishnan
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
- Department of Biomedicine, Bharathidasan University India
| | | | | | - J M Meseguer Dueñas
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València Camino de Vera s/n. 46022 Valencia Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Spain
| | - José Luis Gómez Ribelles
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València Camino de Vera s/n. 46022 Valencia Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Spain
| | | | | | - José Antonio Gómez-Tejedor
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València Camino de Vera s/n. 46022 Valencia Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Spain
| | - Mettu Srinivas Reddy
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
| | | | - Mohamed Rela
- National Foundation for Liver Research, Global Hospitals & Health City Chennai India
| | | |
Collapse
|
243
|
Vázquez N, Sánchez-Arévalo F, Maciel-Cerda A, Garnica-Palafox I, Ontiveros-Tlachi R, Chaires-Rosas C, Piñón-Zarate G, Herrera-Enríquez M, Hautefeuille M, Vera-Graziano R, Castell-Rodríguez A. Influence of the PLGA/gelatin ratio on the physical, chemical and biological properties of electrospun scaffolds for wound dressings. ACTA ACUST UNITED AC 2019; 14:045006. [PMID: 30959495 DOI: 10.1088/1748-605x/ab1741] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chronic wounds are a global health problem, and their treatments are difficult and long lasting. The development of medical devices through tissue engineering has been conducted to heal this type of wound. In this study, it was demonstrated that the combination of natural and synthetic polymers, such as poly (D-L lactide-co-glycolide) (PLGA) and gelatin (Ge), were useful for constructing scaffolds for wound healing. The aim of this study was to evaluate the influence of different PLGA/gelatin ratios (9:1, 7:3 and 5:5 (v/v)) on the physical, chemical and biological properties of electrospun scaffolds for wound dressings. These PLGA/Ge scaffolds had randomly oriented fibers with smooth surfaces and exhibited distances between fibers of less than 10 μm. The 7:3 and 5:5 PLGA/Ge scaffolds showed higher swelling, hydrophilicity and degradation rates than pure PLGA and 9:1 (v/v) PLGA/Ge scaffolds. Young's moduli of the scaffolds were 72 ± 10, 48 ± 6, 58 ± 6 and 6 ± 1 MPa for the pure PLGA scaffold and the 9:1, 7:3 and 5:5 (v/v) PLGA/Ge scaffolds, respectively. Mesenchymal stem cells (MSCs) seeded on all the PLGA/Ge scaffolds were viable, and the cells were attached to the fibers at the different analyzed timepoints. The most significant proliferation rate was observed for cells on the 7:3 PLGA/Ge scaffolds. Biocompatibility analysis showed that all the scaffolds produced inflammation at the first week postimplantation; however, the 7:3 and 5:5 (v/v) PLGA/Ge scaffolds were degraded completely, and there was no inflammatory reaction observed at the fourth week after implantation. In contrast, the 9:1 PLGA/Ge scaffolds persisted in the tissue for more than four weeks; however, at the eighth week, no traces of the scaffolds were found. In conclusion, the scaffolds with the 7:3 PLGA/Ge ratio showed suitable physical, chemical and biological properties for applications in chronic wound treatments.
Collapse
Affiliation(s)
- Nadia Vázquez
- Facultad de Medicina, UNAM, Ciudad de México, C.P. 04510, México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Duarte Campos DF, Rohde M, Ross M, Anvari P, Blaeser A, Vogt M, Panfil C, Yam GH, Mehta JS, Fischer H, Walter P, Fuest M. Corneal bioprinting utilizing collagen‐based bioinks and primary human keratocytes. J Biomed Mater Res A 2019; 107:1945-1953. [DOI: 10.1002/jbm.a.36702] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Daniela F. Duarte Campos
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University Hospital Aachen Germany
| | - Malena Rohde
- Department of OphthalmologyRWTH Aachen University Hospital Aachen Germany
| | - Mitchell Ross
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University Hospital Aachen Germany
- Department of Chemical EngineeringMcMaster University Hamilton Canada
| | - Parham Anvari
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University Hospital Aachen Germany
| | - Andreas Blaeser
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University Hospital Aachen Germany
- Medical Textiles and Biofabrication, Institute for Textile Technology (ITA)RWTH Aachen University Aachen Germany
| | - Michael Vogt
- Interdisciplinary Center for Clinical Research IZKFRWTH Aachen University Hospital Aachen Germany
| | - Claudia Panfil
- Aachen Center for Technology Transfer in Ophthalmology (ACTO) Aachen Germany
| | - Gary Hin‐Fai Yam
- Tissue Engineering and Stem Cell GroupSingapore Eye Research Institute Singapore Singapore
| | - Jodhbir S. Mehta
- Tissue Engineering and Stem Cell GroupSingapore Eye Research Institute Singapore Singapore
- Singapore National Eye Centre Singapore Singapore
| | - Horst Fischer
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University Hospital Aachen Germany
| | - Peter Walter
- Department of OphthalmologyRWTH Aachen University Hospital Aachen Germany
| | - Matthias Fuest
- Department of OphthalmologyRWTH Aachen University Hospital Aachen Germany
| |
Collapse
|
245
|
Khatami N, Khoshfetrat AB, Khaksar M, Zamani ARN, Rahbarghazi R. Collagen‐alginate‐nano‐silica microspheres improved the osteogenic potential of human osteoblast‐like MG‐63 cells. J Cell Biochem 2019; 120:15069-15082. [DOI: 10.1002/jcb.28768] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Neda Khatami
- Chemical Engineering Faculty Sahand University of Technology Tabriz Iran
| | | | - Majid Khaksar
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
| | | | - Reza Rahbarghazi
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
246
|
Pitingolo G, Riaud A, Nastruzzi C, Taly V. Gelatin-Coated Microfluidic Channels for 3D Microtissue Formation: On-Chip Production and Characterization. MICROMACHINES 2019; 10:E265. [PMID: 31010232 PMCID: PMC6523541 DOI: 10.3390/mi10040265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/02/2019] [Accepted: 04/09/2019] [Indexed: 12/23/2022]
Abstract
Traditional two-dimensional (2D) cell culture models are limited in their ability to reproduce human structures and functions. On the contrary, three-dimensional (3D) microtissues have the potential to permit the development of new cell-based assays as advanced in vitro models to test new drugs. Here, we report the use of a dehydrated gelatin film to promote tumor cells aggregation and 3D microtissue formation. The simple and stable gelatin coating represents an alternative to conventional and expensive materials like type I collagen, hyaluronic acid, or matrigel. The gelatin coating is biocompatible with several culture formats including microfluidic chips, as well as standard micro-well plates. It also enables long-term 3D cell culture and in situ monitoring of live/dead assays.
Collapse
Affiliation(s)
- Gabriele Pitingolo
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe Labellisée Ligue Nationale Contre le Cancer, 75005 Paris, France.
| | - Antoine Riaud
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe Labellisée Ligue Nationale Contre le Cancer, 75005 Paris, France.
| | - Claudio Nastruzzi
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Ferrara, 44121 Ferrara, Italy.
| | - Valerie Taly
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe Labellisée Ligue Nationale Contre le Cancer, 75005 Paris, France.
| |
Collapse
|
247
|
Pazarçeviren AE, Evis Z, Keskin D, Tezcaner A. Resorbable PCEC/gelatin-bismuth doped bioglass-graphene oxide bilayer membranes for guided bone regeneration. Biomed Mater 2019; 14:035018. [DOI: 10.1088/1748-605x/ab007b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
248
|
Ardila DC, Tamimi E, Doetschman T, Wagner WR, Vande Geest JP. Modulating smooth muscle cell response by the release of TGFβ2 from tubular scaffolds for vascular tissue engineering. J Control Release 2019. [PMID: 30797003 DOI: 10.1016/j.jconrel.2019.02.0241016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Tissue engineering has gained considerable attention in the development of small diameter tissue engineered vascular grafts (TEVGs) for treating coronary heart disease. A properly designed acellular and biodegradable TEVG must encourage the infiltration and growth of vascular smooth muscle cells (SMCs). Our group has previously shown that increasing levels of TGFβ2 can differentially modulate SMC migration and proliferation. In this study, tubular electrospun scaffolds loaded with TGFβ2 were fabricated using various ratios of gelatin/polycaprolactone (PCL), resulting in scaffolds with porous nano-woven architecture suitable for tissue ingrowth. Scaffold morphology, degradation rate, TGβ2 release kinetics, and bioactivity were assessed. TGFβ2 was successfully integrated into the electrospun biomaterial that resulted in a differential release profile depending on the gelatin/PCL ratio over the course of 42 days. Higher TGFβ2 elution was obtained in scaffolds with higher gelatin content, which may be related to the biodegradation of gelatin in culture media. The biological activity of the released TGFβ2 was evaluated by its ability to affect SMC proliferation as a function of its concentration. SMCs seeded on TGFβ2-loaded scaffolds also showed higher densities and infiltration after 5 days in culture as compared to scaffolds without TGFβ2. Our results demonstrate that the ratio of synthetic and natural polymers in electrospun blends can be used to tune the release of TGFβ2. This method can be used to intelligently modulate the SMC response in gelatin/PCL scaffolds making the TGFβ2-loaded conduits attractive for cardiovascular tissue engineering applications.
Collapse
Affiliation(s)
- D C Ardila
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - E Tamimi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - T Doetschman
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA; BIO5 Institute, The University of Arizona, Tucson, AZ 85724, USA
| | - W R Wagner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - J P Vande Geest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
249
|
Alves P, Santos M, Mendes S, P Miguel S, D de Sá K, S D Cabral C, J Correia I, Ferreira P. Photocrosslinkable Nanofibrous Asymmetric Membrane Designed for Wound Dressing. Polymers (Basel) 2019; 11:E653. [PMID: 30974796 PMCID: PMC6523099 DOI: 10.3390/polym11040653] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/21/2019] [Accepted: 04/08/2019] [Indexed: 12/17/2022] Open
Abstract
Recently, the biomedical scientists who are working in the skin regeneration area have proposed asymmetric membranes as ideal wound dressings, since they are able to reproduce both layers of skin and improve the healing process as well as make it less painful. Herein, an electrospinning technique was used to produce new asymmetric membranes. The protective layer was composed of a blending solution between polycaprolactone and polylactic acid, whereas the underlying layer was comprised of methacrylated gelatin and chitosan. The chemical/physical properties, the in vitro hemo- and biocompatibility of the nanofibrous membranes were evaluated. The results obtained reveal that the produced membranes exhibited a wettability able to provide a moist environment at wound site. Moreover, the membranes' hemocompatibility and fibroblast cell adhesion, spreading and proliferation at the surface of the membranes were also noticed in the in vitro assays. Such results highlight the suitability of these asymmetric membranes for wound dressing applications.
Collapse
Affiliation(s)
- Patrícia Alves
- CIEPQPF, Department of Chemical Engineering, Universidade de Coimbra, P-3030 790 Coimbra, Portugal.
| | - Marta Santos
- CIEPQPF, Department of Chemical Engineering, Universidade de Coimbra, P-3030 790 Coimbra, Portugal.
| | - Sabrina Mendes
- CIEPQPF, Department of Chemical Engineering, Universidade de Coimbra, P-3030 790 Coimbra, Portugal.
| | - Sónia P Miguel
- CICS-UBI, Health Sciences Research Center, Universidade da Beira Interior, P-6200 506 Covilhã, Portugal.
| | - Kevin D de Sá
- CICS-UBI, Health Sciences Research Center, Universidade da Beira Interior, P-6200 506 Covilhã, Portugal.
| | - Cátia S D Cabral
- CICS-UBI, Health Sciences Research Center, Universidade da Beira Interior, P-6200 506 Covilhã, Portugal.
| | - Ilídio J Correia
- CIEPQPF, Department of Chemical Engineering, Universidade de Coimbra, P-3030 790 Coimbra, Portugal.
- CICS-UBI, Health Sciences Research Center, Universidade da Beira Interior, P-6200 506 Covilhã, Portugal.
| | - Paula Ferreira
- CIEPQPF, Department of Chemical Engineering, Universidade de Coimbra, P-3030 790 Coimbra, Portugal.
| |
Collapse
|
250
|
Ardila DC, Tamimi E, Doetschman T, Wagner WR, Vande Geest JP. Modulating smooth muscle cell response by the release of TGFβ2 from tubular scaffolds for vascular tissue engineering. J Control Release 2019; 299:44-52. [PMID: 30797003 PMCID: PMC6430660 DOI: 10.1016/j.jconrel.2019.02.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/25/2019] [Accepted: 02/19/2019] [Indexed: 01/01/2023]
Abstract
Tissue engineering has gained considerable attention in the development of small diameter tissue engineered vascular grafts (TEVGs) for treating coronary heart disease. A properly designed acellular and biodegradable TEVG must encourage the infiltration and growth of vascular smooth muscle cells (SMCs). Our group has previously shown that increasing levels of TGFβ2 can differentially modulate SMC migration and proliferation. In this study, tubular electrospun scaffolds loaded with TGFβ2 were fabricated using various ratios of gelatin/polycaprolactone (PCL), resulting in scaffolds with porous nano-woven architecture suitable for tissue ingrowth. Scaffold morphology, degradation rate, TGβ2 release kinetics, and bioactivity were assessed. TGFβ2 was successfully integrated into the electrospun biomaterial that resulted in a differential release profile depending on the gelatin/PCL ratio over the course of 42 days. Higher TGFβ2 elution was obtained in scaffolds with higher gelatin content, which may be related to the biodegradation of gelatin in culture media. The biological activity of the released TGFβ2 was evaluated by its ability to affect SMC proliferation as a function of its concentration. SMCs seeded on TGFβ2-loaded scaffolds also showed higher densities and infiltration after 5 days in culture as compared to scaffolds without TGFβ2. Our results demonstrate that the ratio of synthetic and natural polymers in electrospun blends can be used to tune the release of TGFβ2. This method can be used to intelligently modulate the SMC response in gelatin/PCL scaffolds making the TGFβ2-loaded conduits attractive for cardiovascular tissue engineering applications.
Collapse
Affiliation(s)
- D C Ardila
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - E Tamimi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - T Doetschman
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA; BIO5 Institute, The University of Arizona, Tucson, AZ 85724, USA
| | - W R Wagner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - J P Vande Geest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|