151
|
Paul MD, Rainwater R, Zuo Y, Gu L, Hristova K. Probing Membrane Protein Association Using Concentration‐Dependent Number and Brightness. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202010049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Michael D. Paul
- Program in Molecular Biophysics Johns Hopkins University Baltimore MD 21218 USA
| | - Randall Rainwater
- Department of Materials Science and Engineering and Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Yi Zuo
- Department of Materials Science and Engineering and Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Luo Gu
- Department of Materials Science and Engineering and Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Kalina Hristova
- Department of Materials Science and Engineering and Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Program in Molecular Biophysics Johns Hopkins University Baltimore MD 21218 USA
| |
Collapse
|
152
|
Flores-Torres S, Peza-Chavez O, Kuasne H, Munguia-Lopez JG, Kort-Mascort J, Ferri L, Jiang T, Rajadurai CV, Park M, Sangwan V, Kinsella JM. Alginate-gelatin-Matrigel hydrogels enable the development and multigenerational passaging of patient-derived 3D bioprinted cancer spheroid models. Biofabrication 2021; 13. [PMID: 33440351 DOI: 10.1088/1758-5090/abdb87] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022]
Abstract
Hydrogels consisting of controlled fractions of alginate, gelatin, and Matrigel enable the development of patient-derived bioprinted tissue models that support cancer spheroid growth and expansion. These engineered models can be dissociated to be then reintroduced to new hydrogel solutions and subsequently reprinted to generate multigenerational models. The process of harvesting cells from 3D bioprinted models is possible by chelating the ions that crosslink alginate, causing the gel to weaken. Inclusion of the gelatin and Matrigel fractions to the hydrogel increases the bioactivity by providing cell-matrix binding sites and promoting cross-talk between cancer cells and their microenvironment. Here we show that immortalized triple-negative breast cancer cells (MDA-MB-231) and patient-derived gastric adenocarcinoma cells can be reprinted for at least three 21 d culture cycles following bioprinting in the alginate/gelatin/Matrigel hydrogels. Our drug testing results suggest that our 3D bioprinted model can also be used to recapitulatein vivopatient drug response. Furthermore, our results show that iterative bioprinting techniques coupled with alginate biomaterials can be used to maintain and expand patient-derived cancer spheroid cultures for extended periods without compromising cell viability, altering division rates, or disrupting cancer spheroid formation.
Collapse
Affiliation(s)
| | - Omar Peza-Chavez
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Hellen Kuasne
- Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Jose G Munguia-Lopez
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.,Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | | | - Lorenzo Ferri
- Department of Surgery, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Tao Jiang
- Department of Intelligent Machinery and Instrument, College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan, People's Republic of China
| | - Charles V Rajadurai
- Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada.,Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Veena Sangwan
- Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Joseph M Kinsella
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
153
|
Chen Y, Hu Z, Zhao D, Zhou K, Huang Z, Zhao W, Yang X, Gao C, Cao Y, Hsu Y, Chang W, Wei Z, Liu X. Self-Assembled Hexagonal Superparamagnetic Cone Structures for Fabrication of Cell Cluster Arrays. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10667-10673. [PMID: 33646740 DOI: 10.1021/acsami.0c17890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In this study, we demonstrated that arrays of cell clusters can be fabricated by self-assembled hexagonal superparamagnetic cone structures. When a strong out-of-plane magnetic field was applied to the ferrofluid on a glass substrate, it will induce the magnetic poles on the upper/lower surfaces of the continuous ferrofluid to increase the magnetostatic energy. The ferrofluid will then experience hydrodynamic instability and be split into small droplets with cone structures because of the compromising surface tension energy and magnetostatic energy to minimize the system's total energy. Furthermore, the ferrofluid cones were orderly self-assembled into hexagonal arrays to reach the lowest energy state. After dehydration of these liquid cones to form solid cones, polydimethylsiloxane was cast to fix the arrangement of hexagonal superparamagnetic cone structures and prevent the leakage of magnetic nanoparticles. The U-343 human neuronal glioblastoma cells were labeled with magnetic nanoparticles through endocytosis in co-culture with a ferrofluid. The number of magnetic nanoparticles internalized was (4.2 ± 0.84) × 106 per cell by the cell magnetophoresis analysis. These magnetically labeled cells were attracted and captured by hexagonal superparamagnetic cone structures to form cell cluster arrays. As a function of the solid cone size, the number of cells captured by each hexagonal superparamagnetic cone structure was increased from 48 to 126 under a 2000 G out-of-plane magnetic field. The local magnetic field gradient of the hexagonal superparamagnetic cone was 117.0-140.9 G/mm from the cell magnetophoresis. When an external magnetic field was applied, we observed that the number of protrusions of the cell edge decreased from the fluorescence images. It showed that the local magnetic field gradient caused by the hexagonal superparamagnetic cones restricted the cell growth and migration.
Collapse
Affiliation(s)
- Yinling Chen
- School of Mechanics and Engineering Science, Zhengzhou University, Zhengzhou 450001, China
- Graduate School of Science and Technology, Shinshu University, Nagano 390-8621, Japan
| | - Zhixin Hu
- School of Mechanics and Engineering Science, Zhengzhou University, Zhengzhou 450001, China
| | - Dongyang Zhao
- School of Mechanics and Engineering Science, Zhengzhou University, Zhengzhou 450001, China
| | - Kejia Zhou
- School of Mechanics and Engineering Science, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenyu Huang
- Division of Cardiology, Johns Hopkins Hospital, Baltimore, Maryland 21287-0010, United States
| | - Wuduo Zhao
- Center of Advance Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaonan Yang
- School of Information Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Chaojun Gao
- School of Physics and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Yangjie Cao
- School of Software & Hanwei Institute of Internet of Things, Zhengzhou University, Zhengzhou 450001, China
| | - Yenya Hsu
- School of Mechanics and Engineering Science, Zhengzhou University, Zhengzhou 450001, China
| | - Weijen Chang
- Department of Biology, Hamilton College, Clinton, New York 13323-1218, United States
| | - Zonhan Wei
- School of Mechanics and Engineering Science, Zhengzhou University, Zhengzhou 450001, China
- School of Information Engineering, Zhengzhou University, Zhengzhou 450001, China
- School of Software & Hanwei Institute of Internet of Things, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaoxi Liu
- Graduate School of Science and Technology, Shinshu University, Nagano 390-8621, Japan
| |
Collapse
|
154
|
Controlled-release of free bacteriophage nanoparticles from 3D-plotted hydrogel fibrous structure as potential antibacterial wound dressing. J Control Release 2021; 331:154-163. [DOI: 10.1016/j.jconrel.2021.01.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/03/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022]
|
155
|
Saska S, Pilatti L, Blay A, Shibli JA. Bioresorbable Polymers: Advanced Materials and 4D Printing for Tissue Engineering. Polymers (Basel) 2021; 13:563. [PMID: 33668617 PMCID: PMC7918883 DOI: 10.3390/polym13040563] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 01/10/2023] Open
Abstract
Three-dimensional (3D) printing is a valuable tool in the production of complexes structures with specific shapes for tissue engineering. Differently from native tissues, the printed structures are static and do not transform their shape in response to different environment changes. Stimuli-responsive biocompatible materials have emerged in the biomedical field due to the ability of responding to other stimuli (physical, chemical, and/or biological), resulting in microstructures modifications. Four-dimensional (4D) printing arises as a new technology that implements dynamic improvements in printed structures using smart materials (stimuli-responsive materials) and/or cells. These dynamic scaffolds enable engineered tissues to undergo morphological changes in a pre-planned way. Stimuli-responsive polymeric hydrogels are the most promising material for 4D bio-fabrication because they produce a biocompatible and bioresorbable 3D shape environment similar to the extracellular matrix and allow deposition of cells on the scaffold surface as well as in the inside. Subsequently, this review presents different bioresorbable advanced polymers and discusses its use in 4D printing for tissue engineering applications.
Collapse
Affiliation(s)
- Sybele Saska
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
| | - Livia Pilatti
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
| | - Alberto Blay
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
| | - Jamil Awad Shibli
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos, Sao Paulo 07023-070, Brazil
| |
Collapse
|
156
|
Łabowska MB, Cierluk K, Jankowska AM, Kulbacka J, Detyna J, Michalak I. A Review on the Adaption of Alginate-Gelatin Hydrogels for 3D Cultures and Bioprinting. MATERIALS (BASEL, SWITZERLAND) 2021; 14:858. [PMID: 33579053 PMCID: PMC7916803 DOI: 10.3390/ma14040858] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/12/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
Sustaining the vital functions of cells outside the organism requires strictly defined parameters. In order to ensure their optimal growth and development, it is necessary to provide a range of nutrients and regulators. Hydrogels are excellent materials for 3D in vitro cell cultures. Their ability to retain large amounts of liquid, as well as their biocompatibility, soft structures, and mechanical properties similar to these of living tissues, provide appropriate microenvironments that mimic extracellular matrix functions. The wide range of natural and synthetic polymeric materials, as well as the simplicity of their physico-chemical modification, allow the mechanical properties to be adjusted for different requirements. Sodium alginate-based hydrogel is a frequently used material for cell culture. The lack of cell-interactive properties makes this polysaccharide the most often applied in combination with other materials, including gelatin. The combination of both materials increases their biological activity and improves their material properties, making this combination a frequently used material in 3D printing technology. The use of hydrogels as inks in 3D printing allows the accurate manufacturing of scaffolds with complex shapes and geometries. The aim of this paper is to provide an overview of the materials used for 3D cell cultures, which are mainly alginate-gelatin hydrogels, including their properties and potential applications.
Collapse
Affiliation(s)
- Magdalena B. Łabowska
- Department of Mechanics, Materials and Biomedical Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland; (M.B.Ł); (A.M.J.)
| | - Karolina Cierluk
- Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373 Wroclaw, Poland;
| | - Agnieszka M. Jankowska
- Department of Mechanics, Materials and Biomedical Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland; (M.B.Ł); (A.M.J.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Jerzy Detyna
- Department of Mechanics, Materials and Biomedical Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland; (M.B.Ł); (A.M.J.)
| | - Izabela Michalak
- Department of Advanced Material Technologies, Faculty of Chemistry, Wroclaw University of Science and Technology, Smoluchowskiego 25, 50-370 Wroclaw, Poland;
| |
Collapse
|
157
|
Paul MD, Rainwater R, Zuo Y, Gu L, Hristova K. Probing Membrane Protein Association Using Concentration-Dependent Number and Brightness. Angew Chem Int Ed Engl 2021; 60:6503-6508. [PMID: 33351993 DOI: 10.1002/anie.202010049] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/11/2020] [Indexed: 01/13/2023]
Abstract
We introduce concentration-dependent number and brightness (cdN&B), a fluorescence fluctuation technique that can be implemented on a standard confocal microscope and can report on the thermodynamics of membrane protein association in the native plasma membrane. It uses transient transfection to enable measurements of oligomer size as a function of receptor concentration over a broad range, yielding the association constant. We discuss artifacts in cdN&B that are concentration-dependent and can distort the oligomerization curves, and we outline procedures that can correct for them. Using cdN&B, we characterize the association of neuropilin 1 (NRP1), a protein that plays a critical role in the development of the embryonic cardiovascular and nervous systems. We show that NRP1 associates into a tetramer in a concentration-dependent manner, and we quantify the strength of the association. This work demonstrates the utility of cdN&B as a powerful tool in biophysical chemistry.
Collapse
Affiliation(s)
- Michael D Paul
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Randall Rainwater
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yi Zuo
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Luo Gu
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kalina Hristova
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.,Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
158
|
Piras CC, Kay AG, Genever PG, Smith DK. Self-assembled low-molecular-weight gelator injectable microgel beads for delivery of bioactive agents. Chem Sci 2021; 12:3958-3965. [PMID: 34163666 PMCID: PMC8179440 DOI: 10.1039/d0sc06296k] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
We report the preparation of hybrid self-assembled microgel beads by combining the low molecular weight gelator (LMWG) DBS-CONHNH2 and the natural polysaccharide calcium alginate polymer gelator (PG). Microgel formulations based on LMWGs are extremely rare due to the fragility of the self-assembled networks and the difficulty of retaining any imposed shape. Our hybrid beads contain interpenetrated LMWG and PG networks, and are obtained by an emulsion method, allowing the preparation of spherical gel particles of controllable sizes with diameters in the mm or μm range. Microgels based on LMWG/alginate can be easily prepared with reproducible diameters <1 μm (ca. 800 nm). They are stable in water at room temperature for many months, and survive injection through a syringe. The rapid assembly of the LMWG on cooling plays an active role in helping control the diameter of the microgel beads. These LMWG microbeads retained the ability of the parent gel to deliver the bioactive molecule heparin, and in cell culture medium this enhanced the growth of human mesenchymal stem cells. Such microgels may therefore have future applications in tissue repair. This approach to fabricating LMWG microgels is a platform technology, which could potentially be applied to a variety of different functional LMWGs, and hence has wide-ranging potential.
Collapse
Affiliation(s)
- Carmen C Piras
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Alasdair G Kay
- Department of Biology, University of York Heslington York YO10 5DD UK
| | - Paul G Genever
- Department of Biology, University of York Heslington York YO10 5DD UK
| | - David K Smith
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| |
Collapse
|
159
|
Naghieh S, Chen X. Printability–A key issue in extrusion-based bioprinting. J Pharm Anal 2021; 11:564-579. [PMID: 34765269 PMCID: PMC8572712 DOI: 10.1016/j.jpha.2021.02.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/25/2022] Open
Affiliation(s)
- Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
- Corresponding author.
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
- Corresponding author. Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada.
| |
Collapse
|
160
|
Sakalem ME, De Sibio MT, da Costa FADS, de Oliveira M. Historical evolution of spheroids and organoids, and possibilities of use in life sciences and medicine. Biotechnol J 2021; 16:e2000463. [PMID: 33491924 DOI: 10.1002/biot.202000463] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND An impressive percentage of biomedical advances were achieved through animal research and cell culture investigations. For drug testing and disease researches, both animal models and preclinical trials with cell cultures are extremely important, but present some limitations, such as ethical concern and inability of representing complex tissues and organs. 3D cell cultures arise providing a more realistic in vitro representation of tissues and organs. Environment and cell type in 3D cultures can represent in vivo conditions and thus provide accurate data on cell-to-cell interactions, and cultivation techniques are based on a scaffold, usually hydrogel or another polymeric material, or without scaffold, such as suspended microplates, magnetic levitation, and microplates for spheroids with ultra-low fixation coating. PURPOSE AND SCOPE This review aims at presenting an updated summary of the most common 3D cell culture models available, as well as a historical background of their establishment and possible applications. SUMMARY Even though 3D culturing is incapable of replacing other current research types, they will continue to substitute some unnecessary animal experimentation, as well as complement monolayer cultures. CONCLUSION In this aspect, 3D culture emerges as a valuable alternative to the investigation of functional, biochemical, and molecular aspects of human pathologies.
Collapse
Affiliation(s)
| | - Maria Teresa De Sibio
- Department of Internal Clinic, Botucatu Medicine School of the Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Felipe Allan da Silva da Costa
- Department of Bioprocesses and Biotechnology, School of Agricultural Sciences of the Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Miriane de Oliveira
- Department of Internal Clinic, Botucatu Medicine School of the Sao Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
161
|
Xu L, Ma F, Huang J, Frankie Leung KL, Qin C, Lu WW, Guo XE, Tang B. Metformin Hydrochloride Encapsulation by Alginate Strontium Hydrogel for Cartilage Regeneration by Reliving Cellular Senescence. Biomacromolecules 2021; 22:671-680. [PMID: 33486954 DOI: 10.1021/acs.biomac.0c01488] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cartilage lesion is a common tissue defect and is challenging in clinical practice. Trauma-induced cellular senescence could decrease the chondrocyte capability of maintaining cartilage tissue regeneration. A previous investigation showed that, by controlling the cellular senescence, the cartilage regeneration can be significantly accelerated. Based on this finding, we design a novel hydrogel, Alg/MH-Sr, that combines metformin, an established drug for inhibiting senescence, and strontium, an effective anti-inflammatory material for cartilage tissue engineering. A RT-PCR test suggests the significant inhibitory effect of the hydrogel on senescent, apoptotic, oxidative, and inflammatory genes' expression. Histological examinations demonstrate that the Alg/MH-Sr hydrogel accelerated cartilage repairment, and chondrocyte senescence was significantly inhibited. Our study demonstrates that the Alg/MH-Sr hydrogel is effective for cartilage defect treatment and provides a new clue in accelerating tissue repairment by inhibiting the senescence of cells and tissues.
Collapse
Affiliation(s)
- Lei Xu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, People's Republic of China.,Department of Orthopeadics and Traumatology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong 999077, SAR, People's Republic of China.,Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, 10027 New York, United States.,Department of Orthopeadics and Traumatology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guang Dong, People's Republic of China
| | - Fenbo Ma
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, People's Republic of China
| | - Jun Huang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, People's Republic of China
| | - Ka Li Frankie Leung
- Department of Orthopeadics and Traumatology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong 999077, SAR, People's Republic of China
| | - Chenghe Qin
- Department of Orthopeadics and Traumatology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guang Dong, People's Republic of China
| | - William Weijia Lu
- Department of Orthopeadics and Traumatology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong 999077, SAR, People's Republic of China
| | - X Edward Guo
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, 10027 New York, United States
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, People's Republic of China.,Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, SUSTech, Schenzhen, Guangdong, People's Republic of China.,Shenzhen Key Laboratory of Cell Microenvironment, SUSTech, Schenzhen, Guangdong, People's Republic of China
| |
Collapse
|
162
|
Abstract
AbstractAlginate is a polysaccharide of natural origin, which shows outstanding properties of biocompatibility, gel forming ability, non-toxicity, biodegradability and easy to process. Due to these excellent properties of alginate, sodium alginate, a hydrogel form of alginate, oxidized alginate and other alginate based materials are used in various biomedical fields, especially in drug delivery, wound healing and tissue engineering. Alginate can be easily processed as the 3D scaffolding materials which includes hydrogels, microcapsules, microspheres, foams, sponges, and fibers and these alginate based bio-polymeric materials have particularly used in tissue healing, healing of bone injuries, scars, wound, cartilage repair and treatment, new bone regeneration, scaffolds for the cell growth. Alginate can be easily modified and blended by adopting some physical and chemical processes and the new alginate derivative materials obtained have new different structures, functions, and properties having improved mechanical strength, cell affinity and property of gelation. This can be attained due to combination with other different biomaterials, chemical and physical crosslinking, and immobilization of definite ligands (sugar and peptide molecules). Hence alginate, its modified forms, derivative and composite materials are found to be more attractive towards tissue engineering. This article provides a comprehensive outline of properties, structural aspects, and application in tissue engineering.
Collapse
|
163
|
González-Orozco JC, Gaona-Domínguez S, Camacho-Arroyo I. In Vitro Models for Studying Tumor Progression. Methods Mol Biol 2021; 2174:193-206. [PMID: 32813251 DOI: 10.1007/978-1-0716-0759-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Beyond cell proliferation, one of the most outstanding characteristics of the cancerous cells that promotes the tumoral progression is their high capacity to migrate and invade the surrounding healthy tissue. These cellular processes (migration and invasion) are critical steps to metastasis. Metastatic progression of the tumors is often the leading cause of morbidity and mortality in cancer patients. Critical genes and cell signaling pathways involved in cell migration and invasion of tumor cells have been identified, and several clinical efforts to alleviate cancer are focused on them; however, once the tumor has metastasized, it is extremely difficult to stop the progression of very aggressive forms of cancer such as glioblastomas. Therefore, it is crucial to elucidate the specific molecular mechanisms underlying tumor progression. In this chapter, we describe some methods to study tumor progression by assessing migration and cell invasion in 2D and 3D cell culture conditions.
Collapse
Affiliation(s)
- Juan Carlos González-Orozco
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México
| | - Saúl Gaona-Domínguez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México.
| |
Collapse
|
164
|
Bhattarai SR, Saudi S, Khanal S, Aravamudhan S, Rorie CJ, Bhattarai N. Electrodynamic assisted self-assembled fibrous hydrogel microcapsules: a novel 3D in vitro platform for assessment of nanoparticle toxicity. RSC Adv 2021; 11:4921-4934. [PMID: 35424445 PMCID: PMC8694512 DOI: 10.1039/d0ra09189h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/07/2021] [Indexed: 12/24/2022] Open
Abstract
Nanoparticle (NP) toxicity assessment is a critical step in assessing the health impacts of NP exposure to both consumers and occupational workers. In vitro assessment models comprising cells cultured in a two-dimensional tissue culture plate (2D-TCP) are an efficient and cost-effective choice for estimating the safety risks of NPs. However, in vitro culture of cells in 2D-TCPs distorts cell–integrin and cell–cell interactions and is not able to replicate an in vivo phenotype. Three-dimensional (3D) in vitro platforms provide a unique alternative to bridge the gap between traditional 2D in vitro and in vivo models. In this study, novel microcapsules of alginate hydrogel incorporated with natural polymeric nanofibers (chitin nanofibrils) and synthetic polymeric nanofibers poly(lactide-co-glycolide) are designed as a 3D in vitro platform. This study demonstrates for the first time that electrodynamic assisted self-assembled fibrous 3D hydrogel (3D-SAF hydrogel) microcapsules with a size in the range of 300–500 μm in diameter with a Young's modulus of 12.7–42 kPa can be obtained by varying the amount of nanofibers in the hydrogel precursor solutions. The 3D-SAF microcapsules were found to mimic the in vivo cellular microenvironment for cells to grow, as evaluated using A549 cells. Higher cellular spreading and prolonged proliferation of A549 cells were observed in 3D-SAF microcapsules compared to control microcapsules without the nanofibers. The 3D-SAF microcapsule integrated well plate was used to assess the toxicity of model NPs, e.g., Al2O3 and ZnO. The toxicity levels of the model NPs were found to be dependent on the chemistry of the NPs and their physical agglomeration in the test media. Our results demonstrate that 3D-SAF microcapsules with an in vivo mimicking microenvironment can be developed as a physiologically relevant platform for high-throughput toxicity screening of NPs or pharmaceutical drugs. Electrohydrodynamic-assisted fabrication of novel nano-net-nanofibrous 3D-SAF hydrogel microcapsules leads to them having tunable mechanical and cell adhesive properties that are applicable to diverse biomedical fields.![]()
Collapse
Affiliation(s)
- Shanta R. Bhattarai
- Department of Biology
- North Carolina A&T State University
- Greensboro
- USA
- Department of Biological Science
| | - Sheikh Saudi
- Department of Nanoengineering
- Joint School of Nanoscience and Nanoengineering
- North Carolina A&T State University
- Greensboro
- USA
| | - Shalil Khanal
- Department of Applied Science and Technology
- North Carolina A&T State University
- Greensboro
- USA
| | - Shyam Aravamudhan
- Department of Nanoengineering
- Joint School of Nanoscience and Nanoengineering
- North Carolina A&T State University
- Greensboro
- USA
| | - Checo J. Rorie
- Department of Biology
- North Carolina A&T State University
- Greensboro
- USA
| | - Narayan Bhattarai
- Department of Chemical, Biological, and Bioengineering
- North Carolina A & T State University
- Greensboro
- USA
| |
Collapse
|
165
|
Zuniga K, Gadde M, Scheftel J, Senecal K, Cressman E, Van Dyke M, Rylander MN. Collagen/kerateine multi-protein hydrogels as a thermally stable extracellular matrix for 3D in vitro models. Int J Hyperthermia 2021; 38:830-845. [PMID: 34058945 PMCID: PMC10523628 DOI: 10.1080/02656736.2021.1930202] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/16/2021] [Accepted: 05/08/2021] [Indexed: 12/30/2022] Open
Abstract
Objective: To determine whether the addition of kerateine (reduced keratin) in rat tail collagen type I hydrogels increases thermal stability and changes material properties and supports cell growth for use in cellular hyperthermia studies for tumor treatment.Methods: Collagen type I extracted from rat tail tendon was combined with kerateine extracted from human hair fibers. Thermal, mechanical, and biocompatibility properties and cell behavior was assessed and compared to 100% collagen type I hydrogels to demonstrate their utility as a tissue model for 3D in vitro testing.Results: A combination (i.e., containing both collagen 'C/KNT') hydrogel was more thermally stable than pure collagen hydrogels and resisted thermal degradation when incubated at a hyperthermic temperature of 47°C for heating durations up to 60 min with a higher melting temperature measured by DSC. An increase in the storage modulus was only observed with an increased collagen concentration rather than an increased KTN concentration; however, a change in ECM structure was observed with greater fiber alignment and width with an increase in KTN concentration. The C/KTN hydrogels, specifically 50/50 C/KTN hydrogels, also supported the growth and of fibroblasts and MDA-MB-231 breast cancer cells similar to those seeded in 100% collagen hydrogels.Conclusion: This multi-protein C/KTN hydrogel shows promise for future studies involving thermal stress studies without compromising the 3D ECM environment or cell growth.
Collapse
Affiliation(s)
- Kameel Zuniga
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Manasa Gadde
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Jacob Scheftel
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Kris Senecal
- Natick Soldier Center, U.S. Army Soldier and Biological Chemical Command, Natick, MA, USA
| | - Erik Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Van Dyke
- College of Biomedical Engineering, The University of Arizona, Tucson, AZ, USA
| | - Marissa Nichole Rylander
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
166
|
Miranda RG, Ferraz ERA, Pereira LC, Dorta DJ. Immunocytochemistry Analysis of HepG2 Cell 3D Culture Encapsulated as Spheroids in Alginate Beads. Methods Mol Biol 2021; 2240:197-206. [PMID: 33423235 DOI: 10.1007/978-1-0716-1091-6_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
3D Cell culture is an alternative to animal use in many drug development and toxicity studies. The 3D cell culture can mimic and reproduce the original tissue microenvironment, morphology, and mechanical and physiological characteristics, to provide a more realistic and reliable response as compared to two-dimensional cultures. 3D cell culture encapsulated in alginate beads is a very simple and relatively inexpensive tool that is easy to handle and to maintain. The alginate beads function as a scaffold that imprisons cells and allows 3D cell growth, to generate spheroids that can have greater genic expression and cell-cell communication as a nano or microtissue. The HepG2 cell line is a human hepatocellular carcinoma cell derivative. HepG2 cells preserve several of the characteristics of hepatocytes and are therefore often used in toxicity studies. Here, we describe HepG2 cell encapsulation in alginate beads and analyze the resulting spheroids formed within the alginate beads by immunocytochemistry, by staining a certain structure with a specific antibody coupled with a fluorophore. This method preserves the beads and enables cell analysis by confocal microscopy.
Collapse
Affiliation(s)
- Raul Ghiraldelli Miranda
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Lilian Cristina Pereira
- Department of Bioprocesses and Biotechnology, Faculty of Agronomic Sciences of Botucatu, São Paulo State University, Botucatu, SP, Brazil
- Center for Evaluation of Environmental Impact on Human Health (TOXICAM), Botucatu, São Paulo, Brazil
| | - Daniel Junqueira Dorta
- FFCLRP-USP, Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
- Instituto Nacional de Tecnologias Alternativas de Detecção, Avaliação Toxicologicae Remoção de Micropututantes e Radioativos (INCT-DATREM), Unesp, Instituto de Química, Araraquara, SP, Brazil.
| |
Collapse
|
167
|
Enck K, Tamburrini R, Deborah C, Gazia C, Jost A, Khalil F, Alwan A, Orlando G, Opara EC. Effect of alginate matrix engineered to mimic the pancreatic microenvironment on encapsulated islet function. Biotechnol Bioeng 2020; 118:1177-1185. [PMID: 33270214 DOI: 10.1002/bit.27641] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/15/2020] [Accepted: 11/21/2020] [Indexed: 12/13/2022]
Abstract
Islet transplantation is emerging as a therapeutic option for type 1 diabetes, albeit, only a small number of patients meeting very stringent criteria are eligible for the treatment because of the side effects of the necessary immunosuppressive therapy and the relatively short time frame of normoglycemia that most patients achieve. The challenge of the immune-suppressive regimen can be overcome through microencapsulation of the islets in a perm-selective coating of alginate microbeads with poly-l-lysine or poly- l-ornithine. In addition to other issues including the nutrient supply challenge of encapsulated islets a critical requirement for these cells has emerged as the need to engineer the microenvironment of the encapsulation matrix to mimic that of the native pancreatic scaffold that houses islet cells. That microenvironment includes biological and mechanical cues that support the viability and function of the cells. In this study, the alginate hydrogel was modified to mimic the pancreatic microenvironment by incorporation of extracellular matrix (ECM). Mechanical and biological changes in the encapsulating alginate matrix were made through stiffness modulation and incorporation of decellularized ECM, respectively. Islets were then encapsulated in this new biomimetic hydrogel and their insulin production was measured after 7 days in vitro. We found that manipulation of the alginate hydrogel matrix to simulate both physical and biological cues for the encapsulated islets enhances the mechanical strength of the encapsulated islet constructs as well as their function. Our data suggest that these modifications have the potential to improve the success rate of encapsulated islet transplantation.
Collapse
Affiliation(s)
- Kevin Enck
- Wake Forest School of Medicine, Virginia Tech School of Biomedical Engineering & Sciences (SBES), Wake Forest University, Winston-Salem, North Carolina.,Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| | - Riccardo Tamburrini
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Chaimov Deborah
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Carlo Gazia
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Alec Jost
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Fatma Khalil
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| | - Abdelrahman Alwan
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| | - Giuseppe Orlando
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Emmanuel C Opara
- Wake Forest School of Medicine, Virginia Tech School of Biomedical Engineering & Sciences (SBES), Wake Forest University, Winston-Salem, North Carolina.,Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| |
Collapse
|
168
|
Palubeckaitė I, Venneker S, Briaire-de Bruijn IH, van den Akker BE, Krol AD, Gelderblom H, Bovée JVMG. Selection of Effective Therapies Using Three-Dimensional in vitro Modeling of Chondrosarcoma. Front Mol Biosci 2020; 7:566291. [PMID: 33425984 PMCID: PMC7793672 DOI: 10.3389/fmolb.2020.566291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/30/2020] [Indexed: 01/04/2023] Open
Abstract
Purpose: Chondrosarcomas are a group of cartilaginous malignant neoplasms characterized by the deposition of chondrogenic extracellular matrix. Surgical resection is currently the only curative treatment option, due to their high resistance to conventional chemotherapy and radiotherapy. Novel therapeutic treatment options may improve outcome. Predominantly used cell line monolayer in vitro models lack in vivo complexity, such as the presence of extracellular matrix, and differing oxygen access. Hence, we aimed to improve pre-clinical chondrosarcoma research by developing an alginate-based 3D cell culture model. Method: An alginate scaffold was applied to generate spheroids of three chondrosarcoma cell lines (CH2879, JJ012, SW1353). Morphological, histological and immunohistochemical assessment of the spheroids were used to characterize the chondrosarcoma model. Presto blue assay, morphological and immunohistochemical assessment were applied to assess spheroid response to a panel of chemotherapeutics and targeted therapies, which was compared to conventional 2D monolayer models. Synergistic effect of doxorubicin and ABT-737 (Bcl-2 inhibitor) was compared between monolayer and spheroid models using excess over Bliss. A 3D colony formation assay was developed for assessment of radiotherapy response. Results: Chondrosarcoma spheroids produced chondrogenic matrix and remained proliferative after 2 weeks of culture. When treated with chemotherapeutics, the spheroids were more resistant than their monolayer counterparts, in line with animal models and clinical data. Moreover, for sapanisertib (mTOR inhibitor) treatment, a recovery in chondrosarcoma growth, previously observed in mice models, was also observed using long-term treatment. Morphological assessment was useful in the case of YM-155 (survivin inhibitor) treatment where a fraction of the spheroids underwent cell death, however a large fraction remained proliferative and unaffected. Synergy was less pronounced in 3D compared to 2D. A 3D clonogenic assay confirmed increased resistance to radiotherapy in 3D chondrosarcoma spheroids. Conclusion: We demonstrate that the chondrosarcoma alginate spheroid model is more representative of chondrosarcoma in vivo and should be used instead of the monolayer model for therapy testing. Improved selection at in vitro stage of therapeutic testing will increase the amount of information available for experimental design of in vivo animal testing and later, clinical stages. This can potentially lead to increased likelihood of approval and success at clinical trials.
Collapse
Affiliation(s)
- Ieva Palubeckaitė
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Sanne Venneker
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Augustinus D Krol
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
169
|
Neethu B, Bhowmick G, Ghangrekar M. Improving performance of microbial fuel cell by enhanced bacterial-anode interaction using sludge immobilized beads with activated carbon. PROCESS SAFETY AND ENVIRONMENTAL PROTECTION 2020; 143:285-292. [DOI: 10.1016/j.psep.2020.06.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
170
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
171
|
Vakili N, Asefnejad A. Titanium coating: introducing an antibacterial and bioactive chitosan-alginate film on titanium by spin coating. ACTA ACUST UNITED AC 2020; 65:621-630. [PMID: 32333647 DOI: 10.1515/bmt-2018-0108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/06/2018] [Indexed: 01/13/2023]
Abstract
Coating of titanium (Ti) implants with biocompatible polymers were performed to improve bone healing. In this study, pure Ti implants were coated via chitosan and alginate by spin coating method at 1000, 4000, and 8000 rpm. The coating layer was cross-linked by calcium chloride. Their chemical structures were analyzed by Fourier transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD) evaluations. The morphology of the created coating was observed by scanning electron microscopy (SEM), and the best uniformity was observed in the prepared coating at 8000 rpm (6093× g) spinal speed. The adhesion strength of the coating layer on the substrate was evaluated by the adhesion pull-off test. Also, the best adhesion strength was achieved at an 8000 rpm (6093× g) coating rate. Bioactivity of the chitosan-alginate coating on Ti sheets was evaluated by soaking the samples in a simulated body fluid (SBF) solution. The apatite formation on prepared Ti sheets was investigated by SEM, XRD, and energy dispersive X-ray spectroscopy (EDS). A higher mineralization appeared on coated samples compared with pure Ti. The antibacterial behavior of the implants was analyzed by bacterial counting against Escherichia coli. The presence of chitosan and alginate on the Ti sheets resulted in a better antibacterial effect. In-vitro experiments, with L929 fibroblast cells, confirmed the biocompatibility of the implants. Coating the Ti implants with chitosan and alginate improved biomineralization and biological behavior of the implant especially at the spinal speed of 8000 rpm (6093× g). These implants can support osteoblast cell adhesion and facilitate bone regeneration.
Collapse
Affiliation(s)
- Nasim Vakili
- Department of Biomedical Engineering, Tehran Science and Research Branch, Islamic Azad University, Tehran 4515-775, Iran
| | - Azadeh Asefnejad
- Department of Biomedical Engineering, Tehran Science and Research Branch, Islamic Azad University, Tehran 4515-775, Iran
| |
Collapse
|
172
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
173
|
Premaratne ID, Toyoda Y, Celie KB, Brown KA, Spector JA. Tissue Engineering Models for the Study of Breast Neoplastic Disease and the Tumor Microenvironment. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:423-442. [DOI: 10.1089/ten.teb.2019.0347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ishani D. Premaratne
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Yoshiko Toyoda
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Karel-Bart Celie
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jason A. Spector
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
174
|
Witte K, de Andrés MC, Wells J, Dalby MJ, Salmeron-Sanchez M, Oreffo ROC. Chondrobags: A high throughput alginate-fibronectin micromass platform for in vitro human cartilage formation. Biofabrication 2020; 12:045034. [DOI: 10.1088/1758-5090/abb653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
175
|
Basu S, Johl R, Pacelli S, Gehrke S, Paul A. Fabricating Tough Interpenetrating Network Cryogels with DNA as the Primary Network for Biomedical Applications. ACS Macro Lett 2020; 9:1230-1236. [PMID: 35638638 DOI: 10.1021/acsmacrolett.0c00448] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This work investigates a sequential strategy to develop DNA-based hydrogel scaffolds with interpenetrating polymeric network. The scaffolds were formed via a two-step procedure. First, a covalently cross-linked DNA-based cryogel was formed by the chemical reaction between DNA strands and a bifunctional cross-linker, polyethylene glycol diepoxide at subzero temperatures. In the second step, alginate chains were absorbed into the preformed macroporous DNA cryogel network, followed by ionic cross-linking with divalent calcium ions. The individual and synergistic effects of covalent and ionic cross-linkings on mechanical and physical properties of the IPN cryogel were tested. The IPN cryogels were able to sustain large deformations higher than 95% of strain under compressive forces without exhibiting any failure. Addition of a physically cross-linked alginate network to the covalently linked DNA cryogel significantly enhanced its toughness and energy dissipation compared to the covalent network alone. The formulated hydrogels also exhibited excellent biocompatibility with human stem cells. Overall, this DNA-based IPN cryogel has the potential to be used as a biomaterial scaffold for a diverse range of tissue engineering applications.
Collapse
Affiliation(s)
- Sayantani Basu
- Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Rea Johl
- Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Settimio Pacelli
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Stevin Gehrke
- Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| |
Collapse
|
176
|
Nilforoushzadeh MA, Khodadadi Yazdi M, Baradaran Ghavami S, Farokhimanesh S, Mohammadi Amirabad L, Zarrintaj P, Saeb MR, Hamblin MR, Zare M, Mozafari M. Mesenchymal Stem Cell Spheroids Embedded in an Injectable Thermosensitive Hydrogel: An In Situ Drug Formation Platform for Accelerated Wound Healing. ACS Biomater Sci Eng 2020; 6:5096-5109. [PMID: 33455261 DOI: 10.1021/acsbiomaterials.0c00988] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ability of mesenchymal stem cells (MSCs) to enhance cutaneous wound healing has been well established. Extensive expansion of cells to reach sufficient cell numbers for regenerating tissues has always limited cell-based therapies. An ingenious solution to address this challenge is to develop a strategy to increase the immunomodulatory effects of MSCs without expanding them. In this study, we employed a simple characteristic of cells. It was observed that an optimized three-dimensional (3D) MSC culture in spheroid forms significantly improved their paracrine effects. An electrospray (ES) encapsulation apparatus was used to encapsulate individual or 3D spheroid MSCs into microscale alginate beads (microbeads). Furthermore, alginate microbeads were embedded in an injectable thermosensitive hydrogel matrix, which gels at skin temperature. The hydrogel fills and seals the wounds cavities, maintains high humidity at the wound area, absorbs exudate, and fixes microbeads, protecting them from direct contact with the harsh wound environment. In vitro investigations revealed that secretion of interleukin 10 (IL-0) and transforming growth factor β1 (TGF-β1) gene was gradually enhanced, providing a delivery platform for prolonged release of bioactive molecules. In vivo study on full-thickness wounds showed granulation and re-epithelialization, only after 7 days. Moreover, increased expression of α-smooth muscle actin (α-SMA) in the first 14 days after treatment ensured wound contraction. Besides, a gradual decrease in α-SMA secretion resulted in reduced scar formation. Well-organized collagen fibrils and high expression of the angiogenesis biomarker CD31 confirmed the promoting effect of the hydrogel on the wound-healing process. The proposed wound-dressing system would potentially be used in scalable and effective cell-based wound therapies.
Collapse
Affiliation(s)
| | | | - Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samila Farokhimanesh
- Department of Biotechnology, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, Oklahoma 74078, United States
| | - Mohammad Reza Saeb
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Mehrak Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
177
|
Mastrorocco A, Cacopardo L, Martino NA, Fanelli D, Camillo F, Ciani E, Roelen BAJ, Ahluwalia A, Dell’Aquila ME. One-step automated bioprinting-based method for cumulus-oocyte complex microencapsulation for 3D in vitro maturation. PLoS One 2020; 15:e0238812. [PMID: 32915922 PMCID: PMC7485809 DOI: 10.1371/journal.pone.0238812] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/23/2020] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional in vitro maturation (3D IVM) is a promising approach to improve IVM efficiency as it could prevent cumulus-oocyte complex (COC) flattening and preserve its structural and functional integrity. Methods reported to date have low reproducibility and validation studies are limited. In this study, a bioprinting based production process for generating microbeads containing a COC (COC-microbeads) was optimized and its validity tested in a large animal model (sheep). Alginate microbeads were produced and characterized for size, shape and stability under culture conditions. COC encapsulation had high efficiency and reproducibility and cumulus integrity was preserved. COC-microbeads underwent IVM, with COCs cultured in standard 2D IVM as controls. After IVM, oocytes were analyzed for nuclear chromatin configuration, bioenergetic/oxidative status and transcriptional activity of genes biomarker of mitochondrial activity (TFAM, ATP6, ATP8) and oocyte developmental competence (KHDC3, NLRP5, OOEP and TLE6). The 3D system supported oocyte nuclear maturation more efficiently than the 2D control (P<0.05). Ooplasmic mitochondrial activity and reactive oxygen species (ROS) generation ability were increased (P<0.05). Up-regulation of TFAM, ATP6 and ATP8 and down-regulation of KHDC3, NLRP5 expression were observed in 3D IVM. In conclusion, the new bioprinting method for producing COC-microbeads has high reproducibility and efficiency. Moreover, 3D IVM improves oocyte nuclear maturation and relevant parameters of oocyte cytoplasmic maturation and could be used for clinical and toxicological applications.
Collapse
Affiliation(s)
- Antonella Mastrorocco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
- * E-mail:
| | | | - Nicola Antonio Martino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Diana Fanelli
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | | - Elena Ciani
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Bernard A. J. Roelen
- Department of Clinical Sciences, Embryology, Anatomy and Physiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Arti Ahluwalia
- Research Centre E. Piaggio, University of Pisa, Pisa, Italy
- Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Maria Elena Dell’Aquila
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
178
|
Jang HY, Shin JY, Oh SH, Byun JH, Lee JH. PCL/HA Hybrid Microspheres for Effective Osteogenic Differentiation and Bone Regeneration. ACS Biomater Sci Eng 2020; 6:5172-5180. [PMID: 33455267 DOI: 10.1021/acsbiomaterials.0c00550] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The purpose of this study is to develop a bioactive bone graft based on polycaprolactone (PCL, synthetic polymer; used in clinical practices as a grafting material for craniofacial bone defects) and hyaluronic acid (HA, bioactive natural polymer; known as a promoting substance for bone regeneration) that would be fabricated by clinically available procedures (mild condition without toxic chemicals) and provide bioactivity for sufficient period, and thus effectively induce bone reconstruction. For this, PCL/HA hybrid microspheres were produced by a spray-precipitation technique using clinically adapted solvents. The HA was stably and evenly entrapped in the PCL/HA hybrid microspheres. It was demonstrated that the PCL/HA hybrid microspheres provide an appropriate environment for proliferation and osteogenic differentiation of human periosteum-derived cells (hPDCs) (in vitro) and allow significantly enhanced bone regeneration (in vivo) compared with PCL microspheres without HA. The PCL/HA hybrid microspheres can be a simple but clinically applicable bioactive bone graft for large-sized bone defects.
Collapse
Affiliation(s)
- Hee Yun Jang
- Department of Advanced Materials, Hannam University, Daejeon 34054, Republic of Korea
| | - Jun Yung Shin
- Department of Advanced Materials, Hannam University, Daejeon 34054, Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea.,Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan 31116, Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jin Ho Lee
- Department of Advanced Materials, Hannam University, Daejeon 34054, Republic of Korea
| |
Collapse
|
179
|
Xing H, Lee H, Luo L, Kyriakides TR. Extracellular matrix-derived biomaterials in engineering cell function. Biotechnol Adv 2020; 42:107421. [PMID: 31381963 PMCID: PMC6995418 DOI: 10.1016/j.biotechadv.2019.107421] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 07/12/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
Extracellular matrix (ECM) derived components are emerging sources for the engineering of biomaterials that are capable of inducing desirable cell-specific responses. This review explores the use of biomaterials derived from naturally occurring ECM proteins and their derivatives in approaches that aim to regulate cell function. Biomaterials addressed are grouped into six categories: purified single ECM proteins, combinations of purified ECM proteins, cell-derived ECM, tissue-derived ECM, diseased and modified ECM, and ECM-polymer coupled biomaterials. Purified ECM proteins serve as a material coating for enhanced cell adhesion and biocompatibility. Cell-derived and tissue-derived ECM, generated by cell isolation and decellularization technologies, can capture the native state of the ECM environment and guide cell migration and alignment patterns as well as stem cell differentiation. We focus primarily on recent advances in the fields of soft tissue, cardiac, and dermal repair, and explore the utilization of ECM proteins as biomaterials to engineer cell responses.
Collapse
Affiliation(s)
- Hao Xing
- Department of Biomedical Engineering, Yale University, United States of America
| | - Hudson Lee
- Department of Molecular Biophysics and Biochemistry, Yale University, United States of America
| | - Lijing Luo
- Department of Pathology, Yale University, United States of America
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, United States of America; Department of Pathology, Yale University, United States of America.
| |
Collapse
|
180
|
Alonzo M, Kumar SA, Allen S, Delgado M, Alvarez-Primo F, Suggs L, Joddar B. Hydrogel scaffolds with elasticity-mimicking embryonic substrates promote cardiac cellular network formation. Prog Biomater 2020; 9:125-137. [PMID: 32978746 PMCID: PMC7544760 DOI: 10.1007/s40204-020-00137-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022] Open
Abstract
Hydrogels are a class of biomaterials used for a wide range of biomedical applications, including as a three-dimensional (3D) scaffold for cell culture that mimics the extracellular matrix (ECM) of native tissues. To understand the role of the ECM in the modulation of cardiac cell function, alginate was used to fabricate crosslinked gels with stiffness values that resembled embryonic (2.66 ± 0.84 kPa), physiologic (8.98 ± 1.29 kPa) and fibrotic (18.27 ± 3.17 kPa) cardiac tissues. The average pore diameter and hydrogel swelling were seen to decrease with increasing substrate stiffness. Cardiomyocytes cultured within soft embryonic gels demonstrated enhanced cell spreading, elongation, and network formation, while a progressive increase in gel stiffness diminished these behaviors. Cell viability decreased with increasing hydrogel stiffness. Furthermore, cells in fibrotic gels showed enhanced protein expression of the characteristic cardiac stress biomarker, Troponin-I, while reduced protein expression of the cardiac gap junction protein, Connexin-43, in comparison to cells within embryonic gels. The results from this study demonstrate the role that 3D substrate stiffness has on cardiac tissue formation and its implications in the development of complex matrix remodeling-based conditions, such as myocardial fibrosis.
Collapse
Affiliation(s)
- Matthew Alonzo
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Shweta Anil Kumar
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Shane Allen
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, Austin, TX, 78712, USA
| | - Monica Delgado
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Fabian Alvarez-Primo
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Laura Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, Austin, TX, 78712, USA
| | - Binata Joddar
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA.
- Department of Metallurgical, Materials and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA.
- Border Biomedical Research Center, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA.
| |
Collapse
|
181
|
Białkowska K, Komorowski P, Bryszewska M, Miłowska K. Spheroids as a Type of Three-Dimensional Cell Cultures-Examples of Methods of Preparation and the Most Important Application. Int J Mol Sci 2020; 21:E6225. [PMID: 32872135 PMCID: PMC7503223 DOI: 10.3390/ijms21176225] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Cell cultures are very important for testing materials and drugs, and in the examination of cell biology and special cell mechanisms. The most popular models of cell culture are two-dimensional (2D) as monolayers, but this does not mimic the natural cell environment. Cells are mostly deprived of cell-cell and cell-extracellular matrix interactions. A much better in vitro model is three-dimensional (3D) culture. Because many cell lines have the ability to self-assemble, one 3D culturing method is to produce spheroids. There are several systems for culturing cells in spheroids, e.g., hanging drop, scaffolds and hydrogels, and these cultures have their applications in drug and nanoparticles testing, and disease modeling. In this paper we would like to present methods of preparation of spheroids in general and emphasize the most important applications.
Collapse
Affiliation(s)
- Kamila Białkowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St. Building D, 90-236 Lodz, Poland; (M.B.); (K.M.)
- Molecular and Nanostructural Biophysics Laboratory, “Bionanopark” Ldt., 114/116 Dubois St., 93-465 Lodz, Poland;
| | - Piotr Komorowski
- Molecular and Nanostructural Biophysics Laboratory, “Bionanopark” Ldt., 114/116 Dubois St., 93-465 Lodz, Poland;
- Department of Biophysics, Institute of Materials Science, Lodz University of Technology, 1/15 Stefanowskiego St., 90-924 Lodz, Poland
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St. Building D, 90-236 Lodz, Poland; (M.B.); (K.M.)
| | - Katarzyna Miłowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St. Building D, 90-236 Lodz, Poland; (M.B.); (K.M.)
| |
Collapse
|
182
|
Hyperpolarized [1- 13C]pyruvate-to-[1- 13C]lactate conversion is rate-limited by monocarboxylate transporter-1 in the plasma membrane. Proc Natl Acad Sci U S A 2020; 117:22378-22389. [PMID: 32839325 DOI: 10.1073/pnas.2003537117] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hyperpolarized [1-13C]pyruvate magnetic resonance spectroscopic imaging (MRSI) is a noninvasive metabolic-imaging modality that probes carbon flux in tissues and infers the state of metabolic reprograming in tumors. Prevailing models attribute elevated hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates in aggressive tumors to enhanced glycolytic flux and lactate dehydrogenase A (LDHA) activity (Warburg effect). By contrast, we find by cross-sectional analysis using genetic and pharmacological tools in mechanistic studies applied to well-defined genetically engineered cell lines and tumors that initial hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates as well as global conversion were highly dependent on and critically rate-limited by the transmembrane influx of [1-13C]pyruvate mediated predominately by monocarboxylate transporter-1 (MCT1). Specifically, in a cell-encapsulated alginate bead model, induced short hairpin (shRNA) knockdown or overexpression of MCT1 quantitatively inhibited or enhanced, respectively, unidirectional pyruvate influxes and [1-13C]pyruvate-to-[1-13C]lactate conversion rates, independent of glycolysis or LDHA activity. Similarly, in tumor models in vivo, hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion was highly dependent on and critically rate-limited by the induced transmembrane influx of [1-13C]pyruvate mediated by MCT1. Thus, hyperpolarized [1-13C]pyruvate MRSI measures primarily MCT1-mediated [1-13C]pyruvate transmembrane influx in vivo, not glycolytic flux or LDHA activity, driving a reinterpretation of this maturing new technology during clinical translation. Indeed, Kaplan-Meier survival analysis for patients with pancreatic, renal, lung, and cervical cancers showed that high-level expression of MCT1 correlated with poor overall survival, and only in selected tumors, coincident with LDHA expression. Thus, hyperpolarized [1-13C]pyruvate MRSI provides a noninvasive functional assessment primarily of MCT1 as a clinical biomarker in relevant patient populations.
Collapse
|
183
|
Synthesis and Characterization of Oxidized Polysaccharides for In Situ Forming Hydrogels. Biomolecules 2020; 10:biom10081185. [PMID: 32824101 PMCID: PMC7464976 DOI: 10.3390/biom10081185] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 12/26/2022] Open
Abstract
Polysaccharides are widely used as building blocks of scaffolds and hydrogels in tissue engineering, which may require their chemical modification to permit crosslinking. The goal of this study was to generate a library of oxidized alginate (oALG) and oxidized hyaluronic acid (oHA) that can be used for in situ gelling hydrogels by covalent reaction between aldehyde groups of the oxidized polysaccharides (oPS) and amino groups of carboxymethyl chitosan (CMC) through imine bond formation. Here, we studied the effect of sodium periodate concentration and reaction time on aldehyde content, molecular weight of derivatives and cytotoxicity of oPS towards 3T3-L1 fibroblasts. It was found that the molecular weights of all oPs decreased with oxidation and that the degree of oxidation was generally higher in oHA than in oALG. Studies showed that only oPs with an oxidation degree above 25% were cytotoxic. Initial studies were also done on the crosslinking of oPs with CMC showing with rheometry that rather soft gels were formed from higher oxidized oPs possessing a moderate cytotoxicity. The results of this study indicate the potential of oALG and oHA for use as in situ gelling hydrogels or inks in bioprinting for application in tissue engineering and controlled release.
Collapse
|
184
|
Li D, Guo B, Liang Q, Liu Y, Zhang L, Hu N, Zhang X, Yang F, Ruan C. Tissue-engineered parathyroid gland and its regulatory secretion of parathyroid hormone. J Tissue Eng Regen Med 2020; 14:1363-1377. [PMID: 32511868 DOI: 10.1002/term.3080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 11/11/2022]
Abstract
Parathyroid glands (PTGs) are important endocrine organs being mainly responsible for the secretion of parathyroid hormone (PTH) to regulate the balance of calcium (Ca) /phosphorus (P) ions in the body. Once PTGs get injured or removed, their resulting defect or loss of PTH secretion should disturb the level of Ca/P in blood, thus damaging other related organs (bone, kidney, etc.) and even causing death. Recently, tissue-engineered PTGs (TE-PTGs) have attracted lots of attention as a potential treatment for the related diseases of PTGs caused by hypoparathyroidism and hyperparathyroidism, including tetany, muscle cramp, nephrolithiasis, nephrocalcinosis, and osteoporosis. Although great progress has been made in the establishment of TE-PTGs with an effective strategy to integrate the key factors of cells and biomaterials, its regulatory secretion of PTH to mimic its natural rhythms in the body remains a huge challenge. This review comprehensively describes an overview of PTGs from physiology and pathology to cytobiology and tissue engineering. The state of the arts in TE-PTGs and the feasible strategies to regulate PTH secretion behaviors are highlighted to provide an important foundation for further investigation.
Collapse
Affiliation(s)
- Duo Li
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Baochun Guo
- Department of Nephrology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, PR China.,Key Laboratory of Shenzhen Renal Diseases, Shenzhen, PR China
| | - Qingfei Liang
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Yunhui Liu
- University of Chinese Academy of Sciences, Beijing, PR China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Lu Zhang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Nan Hu
- Department of Nephrology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, PR China.,Key Laboratory of Shenzhen Renal Diseases, Shenzhen, PR China
| | - Xinzhou Zhang
- Department of Nephrology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, PR China.,Key Laboratory of Shenzhen Renal Diseases, Shenzhen, PR China
| | - Fan Yang
- University of Chinese Academy of Sciences, Beijing, PR China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.,University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
185
|
Kang Y, Datta P, Shanmughapriya S, Ozbolat IT. 3D Bioprinting of Tumor Models for Cancer Research. ACS APPLIED BIO MATERIALS 2020; 3:5552-5573. [DOI: 10.1021/acsabm.0c00791] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Youngnam Kang
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
| | - Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, West Bengal 711103, India
| | - Santhanam Shanmughapriya
- Department of Medicine, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Cellular and Molecular Physiology, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Heart and Vascular Institute, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Ibrahim T. Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
- Biomedical Engineering Department, Penn State University, University Park, Pennsylvania 16802, United States
- Materials Research Institute, Penn State University, University Park, Pennsylvania 16802, United States
- Department of Neurosurgery, Penn State University, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
186
|
Tavakoli J, Wang J, Chuah C, Tang Y. Natural-based Hydrogels: A Journey from Simple to Smart Networks for Medical Examination. Curr Med Chem 2020; 27:2704-2733. [PMID: 31418656 DOI: 10.2174/0929867326666190816125144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Natural hydrogels, due to their unique biological properties, have been used extensively for various medical and clinical examinations that are performed to investigate the signs of disease. Recently, complex-crosslinking strategies improved the mechanical properties and advanced approaches have resulted in the introduction of naturally derived hydrogels that exhibit high biocompatibility, with shape memory and self-healing characteristics. Moreover, the creation of self-assembled natural hydrogels under physiological conditions has provided the opportunity to engineer fine-tuning properties. To highlight recent studies of natural-based hydrogels and their applications for medical investigation, a critical review was undertaken using published papers from the Science Direct database. This review presents different natural-based hydrogels (natural, natural-synthetic hybrid and complex-crosslinked hydrogels), their historical evolution, and recent studies of medical examination applications. The application of natural-based hydrogels in the design and fabrication of biosensors, catheters and medical electrodes, detection of cancer, targeted delivery of imaging compounds (bioimaging) and fabrication of fluorescent bioprobes is summarised here. Without doubt, in future, more useful and practical concepts will be derived to identify natural-based hydrogels for a wide range of clinical examination applications.
Collapse
Affiliation(s)
- Javad Tavakoli
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia.,School of Biomedical Engineering, University of Technology Sydney, Ultimo, 2007 NSW, Australia
| | - Jing Wang
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia.,Key Laboratory of Advanced Textile Composite Materials of Ministry of Education, Institute of Textile Composite, School of Textile, Tianjin Polytechnic University, Tianjin 300387, China
| | - Clarence Chuah
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia
| | - Youhong Tang
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia
| |
Collapse
|
187
|
Mu X, Fitzpatrick V, Kaplan DL. From Silk Spinning to 3D Printing: Polymer Manufacturing using Directed Hierarchical Molecular Assembly. Adv Healthc Mater 2020; 9:e1901552. [PMID: 32109007 PMCID: PMC7415583 DOI: 10.1002/adhm.201901552] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/18/2019] [Indexed: 12/25/2022]
Abstract
Silk spinning offers an evolution-based manufacturing strategy for industrial polymer manufacturing, yet remains largely inaccessible as the manufacturing mechanisms in biological and synthetic systems, especially at the molecular level, are fundamentally different. The appealing characteristics of silk spinning include the sustainable sourcing of the protein material, the all-aqueous processing into fibers, and the unique material properties of silks in various formats. Substantial progress has been made to mimic silk spinning in artificial manufacturing processes, despite the gap between natural and artificial systems. This report emphasizes the universal spinning conditions utilized by both spiders and silkworms to generate silk fibers in nature, as a scientific and technical framework for directing molecular assembly into high-performance structures. The preparation of regenerated silk feedstocks and mimicking native spinning conditions in artificial manufacturing are discussed, as is progress and challenges in fiber spinning and 3D printing of silk-composites. Silk spinning is a biomimetic model for advanced and sustainable artificial polymer manufacturing, offering benefits in biomedical applications for tissue scaffolds and implantable devices.
Collapse
Affiliation(s)
- Xuan Mu
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
188
|
An C, Liu W, Zhang Y, Pang B, Liu H, Zhang Y, Zhang H, Zhang L, Liao H, Ren C, Wang H. Continuous microfluidic encapsulation of single mesenchymal stem cells using alginate microgels as injectable fillers for bone regeneration. Acta Biomater 2020; 111:181-196. [PMID: 32450230 DOI: 10.1016/j.actbio.2020.05.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/13/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
The encapsulation of cells in microscale hydrogels can provide a mimic of a three-dimensional (3D) microenvironment to support cell viability and functions and to protect cells from the environmental stress, which have been widely used in tissue regeneration and cell therapies. Here, a microfluidics-based approach is developed for continuous encapsulation of mesenchymal stem cells (MSCs) at the single-cell level using alginate microgels. This microfluidic technique integrated on-chip encapsulation, gelation, and de-emulsification into a one-step fabrication process, which enables scalable cell encapsulation while retaining the viability and functionality of loaded cells. Remarkably, we observed MSCs encapsulated in Ca-alginate microgels at the single-cell level showed significantly enhanced osteogenesis and accelerated mineralization of the microgels which occurred only after 7 days of induction. Furthermore, MSCs laden in alginate microgels displayed significantly enhanced bone formation compared to MSCs mixed with microgels and acellular microgels in a rat tibial ablation model. To conclude, the current microfluidic technique represents a significant step toward continuous single cell encapsulation, fabrication, and purification. These microgels can boost bone regeneration by providing a controlled osteogenic microenvironment for encapsulated MSCs and facilitate stem cell therapy in the treatment of bone defects in a minimally invasive delivery way. STATEMENT OF SIGNIFICANCE: The biological functions and therapeutic activities of single cells laden in microgels for tissue engineering remains less investigated. Here, we reported a microfluidic-based method for continuous encapsulation of single MSCs with high viability and functionality by integrating on-chip encapsulation, gelation, and de-emulsification into a one-step fabrication process. More importantly, MSCs encapsulated in alginate microgels at the single-cell level showed significantly enhanced osteogenesis, remarkably accelerated mineralization in vitro and bone formation capacity in vivo. Therefore, this single-cell encapsulation technique can facilitate stem cell therapy for bone regeneration and be potentially used in a variety of tissue engineering applications.
Collapse
|
189
|
Piras CC, Mahon CS, Smith DK. Self-Assembled Supramolecular Hybrid Hydrogel Beads Loaded with Silver Nanoparticles for Antimicrobial Applications. Chemistry 2020; 26:8452-8457. [PMID: 32294272 PMCID: PMC7384024 DOI: 10.1002/chem.202001349] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/08/2020] [Indexed: 12/28/2022]
Abstract
This Full Paper reports the formation of silver (Ag) NPs within spatially resolved two-component hydrogel beads, which combine a low-molecular-weight gelator (LMWG) DBS-CONHNH2 and a polymer gelator (PG) calcium alginate. The AgNPs are formed through in situ reduction of AgI , with the resulting nanoparticle-loaded gels being characterised in detail. The antibacterial activity of the nanocomposite gel beads was tested against two drug-resistant bacterial strains, often associated with hospital-acquired infections: vancomycin-resistant Enterococcus faecium (VRE) and Pseudomonas aeruginosa (PA14), and the AgNP-loaded gels showed good antimicrobial properties against both types of bacteria. It is suggested that the gel bead format of these AgNP-loaded hybrid hydrogels makes them promising versatile materials for potential applications in orthopaedics or wound healing.
Collapse
Affiliation(s)
- Carmen C. Piras
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
| | - Clare S. Mahon
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
| | - David K. Smith
- Department of ChemistryUniversity of YorkHeslingtonYorkYO10 5DDUK
| |
Collapse
|
190
|
Lutzweiler G, Ndreu Halili A, Engin Vrana N. The Overview of Porous, Bioactive Scaffolds as Instructive Biomaterials for Tissue Regeneration and Their Clinical Translation. Pharmaceutics 2020; 12:E602. [PMID: 32610440 PMCID: PMC7407612 DOI: 10.3390/pharmaceutics12070602] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Porous scaffolds have been employed for decades in the biomedical field where researchers have been seeking to produce an environment which could approach one of the extracellular matrixes supporting cells in natural tissues. Such three-dimensional systems offer many degrees of freedom to modulate cell activity, ranging from the chemistry of the structure and the architectural properties such as the porosity, the pore, and interconnection size. All these features can be exploited synergistically to tailor the cell-material interactions, and further, the tissue growth within the voids of the scaffold. Herein, an overview of the materials employed to generate porous scaffolds as well as the various techniques that are used to process them is supplied. Furthermore, scaffold parameters which modulate cell behavior are identified under distinct aspects: the architecture of inert scaffolds (i.e., pore and interconnection size, porosity, mechanical properties, etc.) alone on cell functions followed by comparison with bioactive scaffolds to grasp the most relevant features driving tissue regeneration. Finally, in vivo outcomes are highlighted comparing the accordance between in vitro and in vivo results in order to tackle the future translational challenges in tissue repair and regeneration.
Collapse
Affiliation(s)
- Gaëtan Lutzweiler
- Institut National de la Santé et de la Recherche Medicale, UMR_S 1121, 11 rue Humann, 67085 Strasbourg CEDEX, France
| | - Albana Ndreu Halili
- Department of Information Technology, Aleksander Moisiu University, 2001 Durres, Albania;
| | | |
Collapse
|
191
|
Patra T, Gupta MK. Evaluation of sodium alginate for encapsulation-vitrification of testicular Leydig cells. Int J Biol Macromol 2020; 153:128-137. [PMID: 32092420 DOI: 10.1016/j.ijbiomac.2020.02.233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/10/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022]
Abstract
This study reports encapsulation-vitrification of Leydig cells. The Leydig cells were encapsulated in sodium alginate beads of different sizes and cryopreserved by vitrification or slow freezing. Physico-chemical characterization of beads was done by Fourier Transform Infrared Spectroscopy (FTIR), X-Ray Diffraction (XRD), Fluorescence Recovery after Photobleaching (FRAP) and in vitro biodegradation study. Surface morphology of cryopreserved cell-encapsulated beads was evaluated by Environmental Scanning Electron Microscopy (E-SEM), encapsulation efficiency and viability of cells were assessed by Trypan blue assay, mitochondrial activity (MTT assay) and cytoplasmic esterase enzyme activity (FDA assay), respectively. Results showed that vitrification gives better results than slow freezing with respect to surface morphology as well as cell viability of the cell-encapsulated beads (86.94 ± 2.20% vs. 67.94 ± 2.30%; p < 0.05). Encapsulation of cells in small diameter beads (1.8 mm) gave a better cell proliferation rate than large (2.1 mm and 2.7 mm). There was a significant difference in the population doubling time (47.9 ± 1.7 h vs. 67.1 ± 2.5 h) and cell proliferation rate (0.50 ± 0.24 vs. 0.36 ± 0.24 per day) of vitrified-warmed cell encapsulated beads with different diameter (p < 0.05). Encapsualtion in sodium alginate beads is a promising method for cryopreservation of Leydig cells by slow freezing as well as vitrification.
Collapse
Affiliation(s)
- Tanushree Patra
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Mukesh Kumar Gupta
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
192
|
Jose G, Shalumon K, Chen JP. Natural Polymers Based Hydrogels for Cell Culture Applications. Curr Med Chem 2020; 27:2734-2776. [PMID: 31480996 DOI: 10.2174/0929867326666190903113004] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
Abstract
It is well known that the extracellular matrix (ECM) plays a vital role in the growth, survival
and differentiation of cells. Though two-dimensional (2D) materials are generally used as substrates for
the standard in vitro experiments, their mechanical, structural, and compositional characteristics can
alter cell functions drastically. Many scientists reported that cells behave more natively when cultured
in three-dimensional (3D) environments than on 2D substrates, due to the more in vivo-like 3D cell
culture environment that can better mimic the biochemical and mechanical properties of the ECM. In
this regard, water-swollen network polymer-based materials called hydrogels are highly attractive for
developing 3D ECM analogs due to their biocompatibility and hydrophilicity. Since hydrogels can be
tuned and altered systematically, these materials can function actively in a defined culture medium to
support long-term self-renewal of various cells. The physico-chemical and biological properties of the
materials used for developing hydrogel should be tunable in accordance with culture needs. Various
types of hydrogels derived either from natural or synthetic origins are currently being used for cell culture
applications. In this review, we present an overview of various hydrogels based on natural polymers
that can be used for cell culture, irrespective of types of applications. We also explain how each
hydrogel is made, its source, pros and cons in biological applications with a special focus on regenerative
engineering.
Collapse
Affiliation(s)
- Gils Jose
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - K.T. Shalumon
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| |
Collapse
|
193
|
Gao H, Liu C, Wu B, Cui H, Zhao Y, Duan Y, Gao F, Gu Q, Wang H, Li W. Effects of Different Biomaterials and Cellular Status on Testicular Cell Self-Organization. ACTA ACUST UNITED AC 2020; 4:e1900292. [PMID: 32453509 DOI: 10.1002/adbi.201900292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/03/2020] [Indexed: 01/12/2023]
Abstract
A multicellular organism's development is coupled with cellular self-organization, which is regulated by cell-cell interactions and cell-extracellular matrix (ECM) crosstalk. Testicular cells from different species such as mouse, rat, and porcine can self-organize into seminiferous tubules both in vitro and in vivo, but the understanding of the functional role of the ECM during this process is limited. Here, it is shown that mouse testicular cells encapsulated with the biomaterial Matrigel can self-organize into seminiferous tubules with blood-testis barrier (BTB) formation and Leydig cell differentiation. By varying the encapsulation method, a combination of sodium alginate and collagen is used to promote reorganization of seminiferous tubules, which resemble those in vivo. In addition, the self-organization ability of testicular cells declines with advanced cell age, and those germ cells play a pivotal role in this process. These findings will be helpful to understand the self-organization process of testicular cells and provide insights for the reconstruction of testes.
Collapse
Affiliation(s)
- Hui Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hang Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Yan Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yongchao Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Stem cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Colleague of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
194
|
Soleimani A, Fard NZ, Talaei-Khozani T, Bahmanpour S. Epidermal growth factor and three-dimensional scaffolds provide conducive environment for differentiation of mouse embryonic stem cells into oocyte-like cells. Cell Biol Int 2020; 44:1850-1859. [PMID: 32437076 DOI: 10.1002/cbin.11391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 04/07/2020] [Accepted: 05/18/2020] [Indexed: 11/09/2022]
Abstract
Three-dimensional (3D) culture provides a biomimicry of the naive microenvironment that can support cell proliferation, differentiation, and regeneration. Some growth factors, such as epidermal growth factor (EGF), facilitate normal meiosis during oocyte maturation in vivo. In this study, a scaffold-based 3D coculture system using purified alginate was applied to induce oocyte differentiation from mouse embryonic stem cells (mESCs). mESCs were induced to differentiate into oocyte-like cells using embryoid body protocol in the two-dimensional or 3D microenvironment in vitro. To increase the efficiency of the oocyte-like cell differentiation from mESCs, we employed a coculture system using ovarian granulosa cells in the presence or absence of epidermal growth factor (+EGF or -EGF) for 14 days and then the cells were assessed for germ cell differentiation, meiotic progression, and oocyte maturation markers. The cultures exposed to EGF in the alginate-based 3D microenvironment showed the highest level of premeiotic (Oct4 and Mvh), meiotic (Scp1, Scp3, Stra8, and Rec8), and oocyte maturation (Gdf9, Cx37, and Zp2) marker genes (p < .05) in comparison to other groups. According to the gene-expression patterns, we can conclude that alginate-based 3D coculture system provided a highly efficient protocol for oocyte-like cell differentiation from mESCs. The data showed that this culture system along with EGF improved the rate of in vitro oocyte-like cell differentiation.
Collapse
Affiliation(s)
- Azam Soleimani
- Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nehleh Zarei Fard
- Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Bahmanpour
- Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
195
|
Schwieger J, Hamm A, Gepp MM, Schulz A, Hoffmann A, Lenarz T, Scheper V. Alginate-encapsulated brain-derived neurotrophic factor-overexpressing mesenchymal stem cells are a promising drug delivery system for protection of auditory neurons. J Tissue Eng 2020; 11:2041731420911313. [PMID: 32341778 PMCID: PMC7168777 DOI: 10.1177/2041731420911313] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/08/2020] [Indexed: 12/23/2022] Open
Abstract
The cochlear implant outcome is possibly improved by brain-derived neurotrophic factor treatment protecting spiral ganglion neurons. Implantation of genetically modified mesenchymal stem cells may enable the required long-term brain-derived neurotrophic factor administration. Encapsulation of mesenchymal stem cells in ultra-high viscous alginate may protect the mesenchymal stem cells from the recipient’s immune system and prevent their uncontrolled migration. Alginate stability and survival of mesenchymal stem cells in alginate were evaluated. Brain-derived neurotrophic factor production was measured and its protective effect was analyzed in dissociated rat spiral ganglion neuron co-culture. Since the cochlear implant is an active electrode, alginate–mesenchymal stem cell samples were electrically stimulated and alginate stability and mesenchymal stem cell survival were investigated. Stability of ultra-high viscous-alginate and alginate–mesenchymal stem cells was proven. Brain-derived neurotrophic factor production was detectable and spiral ganglion neuron survival, bipolar morphology, and neurite outgrowth were increased. Moderate electrical stimulation did not affect the mesenchymal stem cell survival and their viability was good within the investigated time frame. Local drug delivery by ultra-high viscous-alginate-encapsulated brain-derived neurotrophic factor–overexpressing mesenchymal stem cells is a promising strategy to improve the cochlear implant outcome.
Collapse
Affiliation(s)
- Jana Schwieger
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Anika Hamm
- NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| | - Michael M Gepp
- Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach, Germany.,Fraunhofer Project Center for Stem Cell Process Engineering, Würzburg, Germany
| | - André Schulz
- Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach, Germany
| | - Andrea Hoffmann
- NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Cluster of Excellence Hearing4all, German Research Foundation, Hannover, Germany
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,NIFE-Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany.,Cluster of Excellence Hearing4all, German Research Foundation, Hannover, Germany
| |
Collapse
|
196
|
Abalymov A, Van der Meeren L, Saveleva M, Prikhozhdenko E, Dewettinck K, Parakhonskiy B, Skirtach AG. Cells-Grab-on Particles: A Novel Approach to Control Cell Focal Adhesion on Hybrid Thermally Annealed Hydrogels. ACS Biomater Sci Eng 2020; 6:3933-3944. [DOI: 10.1021/acsbiomaterials.0c00119] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Anatolii Abalymov
- Department of Biotechnology, University of Ghent, Ghent 9000, Belgium
- Remote Controlled Theranostic Systems Lab, Educational Research Institute of Nanostructures and Biosystems, Saratov State University, Saratov 410012, Russia
| | | | - Mariia Saveleva
- Department of Biotechnology, University of Ghent, Ghent 9000, Belgium
- Remote Controlled Theranostic Systems Lab, Educational Research Institute of Nanostructures and Biosystems, Saratov State University, Saratov 410012, Russia
| | - Ekaterina Prikhozhdenko
- Faculty of Nano- and Biomedical Technologies, Saratov State University, Saratov 410012, Russia
| | - Koen Dewettinck
- Department of Food Technology, Safety & Health, University of Gent, Ghent 9000, Belgium
| | | | - Andre G. Skirtach
- Department of Biotechnology, University of Ghent, Ghent 9000, Belgium
| |
Collapse
|
197
|
Abalymov A, Parakhonskiy B, Skirtach AG. Polymer- and Hybrid-Based Biomaterials for Interstitial, Connective, Vascular, Nerve, Visceral and Musculoskeletal Tissue Engineering. Polymers (Basel) 2020; 12:E620. [PMID: 32182751 PMCID: PMC7182904 DOI: 10.3390/polym12030620] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/19/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review, materials based on polymers and hybrids possessing both organic and inorganic contents for repairing or facilitating cell growth in tissue engineering are discussed. Pure polymer based biomaterials are predominantly used to target soft tissues. Stipulated by possibilities of tuning the composition and concentration of their inorganic content, hybrid materials allow to mimic properties of various types of harder tissues. That leads to the concept of "one-matches-all" referring to materials possessing the same polymeric base, but different inorganic content to enable tissue growth and repair, proliferation of cells, and the formation of the ECM (extra cellular matrix). Furthermore, adding drug delivery carriers to coatings and scaffolds designed with such materials brings additional functionality by encapsulating active molecules, antibacterial agents, and growth factors. We discuss here materials and methods of their assembly from a general perspective together with their applications in various tissue engineering sub-areas: interstitial, connective, vascular, nervous, visceral and musculoskeletal tissues. The overall aims of this review are two-fold: (a) to describe the needs and opportunities in the field of bio-medicine, which should be useful for material scientists, and (b) to present capabilities and resources available in the area of materials, which should be of interest for biologists and medical doctors.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | | | - Andre G. Skirtach
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
198
|
Unagolla JM, Jayasuriya AC. Hydrogel-based 3D bioprinting: A comprehensive review on cell-laden hydrogels, bioink formulations, and future perspectives. APPLIED MATERIALS TODAY 2020; 18:100479. [PMID: 32775607 PMCID: PMC7414424 DOI: 10.1016/j.apmt.2019.100479] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Hydrogel plays a vital role in cell-laden three dimensional (3D) bioprinting, whereas those hydrogels mimic the physical and biochemical characteristics of native extracellular matrix (ECM). The complex microenvironment of the ECM does not replicate from the traditional static microenvironment of the hydrogel, but the evolution of the 3D bioprinting facilitates to accommodate the dynamic modulation and spatial heterogeneity of the hydrogel system. Selection of hydrogel for 3D bioprinting depends on the printing techniques including microextrusion, inkjet, laser-assisted printing, and stereolithography. In this review, we specifically cover the 3D printable hydrogels where cells can be encapsulated without significant reduction in the cell viability. The recent research highlights of the most widely used hydrogel materials are elucidated in terms of stability of the hydrogel system, cross-linking method, support cell types and their post-printing cell viability. Also, the techniques used to improve the mechanical and biological properties of the hydrogels, such as adding various organic and inorganic materials and making microchannels, are discussed. Furthermore, the recent advances in vascularized tissue construct and scaffold-free bioprinting as a promising method for vascularization are covered in this review. The recent trends in four-dimensional (4D) bioprinting as a stimuli-responsive formation of new organs, and 3D bioprinting based organ-on-chip systems are also discussed.
Collapse
Affiliation(s)
- Janitha M. Unagolla
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43607, USA
| | - Ambalangodage C. Jayasuriya
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43607, USA
- Department of Orthopedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
199
|
Lee SS, Kim H, Sohn DK, Eom JB, Seo YS, Yoon HM, Choi Y. Indocyanine green-loaded injectable alginate hydrogel as a marker for precision cancer surgery. Quant Imaging Med Surg 2020; 10:779-788. [PMID: 32269936 DOI: 10.21037/qims.2020.02.24] [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: 12/20/2022]
Abstract
Background Accurate identification of tumor sites and boundaries is of paramount importance during minimally invasive surgery. Although laparoscopic resection is being increasingly and widely performed for early gastric and colorectal cancers, the detection of tumors located inside the stomach and intestine is difficult owing to the lack of tactile sensation. Here, we propose the application of an indocyanine green (ICG)-loaded alginate hydrogel system as a fluorescence surgical marker for precise laparoscopic operations. Methods A physical complex of ICG and human serum albumin (HSA) was mixed with sodium alginate to form an injectable hydrogel system. Calcium carbonate and D-gluconic acid (GA) were added to the gel to control its strength and gelation time, respectively. The optimal conditions for the preparation of injectable hydrogels were determined by analyzing the fluorescence spectra and sol-gel transition time of the prepared samples at various concentrations and compositions. Next, the aqueous solutions of ICG, ICG-HSA, and ICG-HSA-loaded alginate were subcutaneously injected into nude mice (three mice per group), and near-infrared (NIR) fluorescence images of the mice (λex. =780 nm, λem. =845 nm) were obtained at different points in time for 8 days. Then, fluorescence intensities at the injection sites, target-to-background ratio, and areas of ICG fluorescence were analyzed. Finally, the potential utility of ICG-HSA-loaded alginate hydrogel as a surgical marker was evaluated in a porcine model. The ICG-HSA-loaded alginate solution was injected into three sites in the submucosal space of the porcine stomach via a catheter. A fluorescent laparoscopic system was installed on the abdomen of the pig 3 days post-injection, and the fluorescence signal generated from the fluorescence surgical marker located inside the stomach was evaluated using the fluorescence laparoscope system (λex. =785 nm, λem. =805 nm). Results The optimal concentration of ICG-HSA complex was determined to be 30 µM, and maximum fluorescence intensity of the complex was obtained at a 1:1 mole ratio of HSA to ICG. The subcutaneous injection of ICG or ICG-HSA solution in mice resulted in the rapid spread of the fluorescence signal around the injection site in 3 h, and a weak fluorescence was detected at the injection site 24 h post-injection. In contrast, the fluorescence detection time was effectively prolonged up to 96 h post-injection in the case of ICG-HSA-loaded alginate gel, while diffusion of the injected ICG from the injection site was effectively prevented. In the laparoscopic operation, injection sites of the hydrogel in porcine stomach could be accurately detected in real time even after 3 days. Conclusions This alginate hydrogel system may be potentially useful as an effective surgical marker in terms of accuracy and persistence for laparoscopic operation.
Collapse
Affiliation(s)
- Seon Sook Lee
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Hyunjin Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Dae Kyung Sohn
- Center for Colorectal Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Joo Beom Eom
- College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Young Seok Seo
- R&D Center, Wontech Co., Ltd., Daejeon, Republic of Korea
| | - Hong Man Yoon
- Center for Gastric Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Yongdoo Choi
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
200
|
Bedell ML, Melchiorri AJ, Aleman J, Skardal A, Mikos AG. A high-throughput approach to compare the biocompatibility of candidate bioink formulations. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2019.e00068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|