1
|
Barrett L, Coopman K. Cell microencapsulation techniques for cancer modelling and drug discovery. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:345-354. [PMID: 38829715 DOI: 10.1080/21691401.2024.2359996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Cell encapsulation into spherical microparticles is a promising bioengineering tool in many fields, including 3D cancer modelling and pre-clinical drug discovery. Cancer microencapsulation models can more accurately reflect the complex solid tumour microenvironment than 2D cell culture and therefore would improve drug discovery efforts. However, these microcapsules, typically in the range of 1 - 5000 µm in diameter, must be carefully designed and amenable to high-throughput production. This review therefore aims to outline important considerations in the design of cancer cell microencapsulation models for drug discovery applications and examine current techniques to produce these. Extrusion (dripping) droplet generation and emulsion-based techniques are highlighted and their suitability to high-throughput drug screening in terms of tumour physiology and ease of scale up is evaluated.
Collapse
Affiliation(s)
- Lisa Barrett
- Department of Chemical Engineering, School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, UK
| | - Karen Coopman
- Department of Chemical Engineering, School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
2
|
Ryoo H, Kimmel H, Rondo E, Underhill GH. Advances in high throughput cell culture technologies for therapeutic screening and biological discovery applications. Bioeng Transl Med 2024; 9:e10627. [PMID: 38818120 PMCID: PMC11135158 DOI: 10.1002/btm2.10627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 06/01/2024] Open
Abstract
Cellular phenotypes and functional responses are modulated by the signals present in their microenvironment, including extracellular matrix (ECM) proteins, tissue mechanical properties, soluble signals and nutrients, and cell-cell interactions. To better recapitulate and analyze these complex signals within the framework of more physiologically relevant culture models, high throughput culture platforms can be transformative. High throughput methodologies enable scientists to extract increasingly robust and broad datasets from individual experiments, screen large numbers of conditions for potential hits, better qualify and predict responses for preclinical applications, and reduce reliance on animal studies. High throughput cell culture systems require uniformity, assay miniaturization, specific target identification, and process simplification. In this review, we detail the various techniques that researchers have used to face these challenges and explore cellular responses in a high throughput manner. We highlight several common approaches including two-dimensional multiwell microplates, microarrays, and microfluidic cell culture systems as well as unencapsulated and encapsulated three-dimensional high throughput cell culture systems, featuring multiwell microplates, micromolds, microwells, microarrays, granular hydrogels, and cell-encapsulated microgels. We also discuss current applications of these high throughput technologies, namely stem cell sourcing, drug discovery and predictive toxicology, and personalized medicine, along with emerging opportunities and future impact areas.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Hannah Kimmel
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Evi Rondo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Gregory H. Underhill
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
3
|
Ye M, Shan Y, Lu B, Luo H, Li B, Zhang Y, Wang Z, Guo Y, Ouyang L, Gu J, Xiong Z, Zhang T. Creating a semi-opened micro-cavity ovary through sacrificial microspheres as an in vitro model for discovering the potential effect of ovarian toxic agents. Bioact Mater 2023; 26:216-230. [PMID: 36936809 PMCID: PMC10017366 DOI: 10.1016/j.bioactmat.2023.02.029] [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: 11/30/2022] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/09/2023] Open
Abstract
The bio-engineered ovary is an essential technology for treating female infertility. Especially the development of relevant in vitro models could be a critical step in a drug study. Herein, we develop a semi-opened culturing system (SOCS) strategy that maintains a 3D structure of follicles during the culture. Based on the SOCS, we further developed micro-cavity ovary (MCO) with mouse follicles by the microsphere-templated technique, where sacrificial gelatin microspheres were mixed with photo-crosslinkable gelatin methacryloyl (GelMA) to engineer a micro-cavity niche for follicle growth. The semi-opened MCO could support the follicle growing to the antral stage, secreting hormones, and ovulating cumulus-oocyte complex out of the MCO without extra manipulation. The MCO-ovulated oocyte exhibits a highly similar transcriptome to the in vivo counterpart (correlation of 0.97) and can be fertilized. Moreover, we found that a high ROS level could affect the cumulus expansion, which may result in anovulation disorder. The damage could be rescued by melatonin, but the end of cumulus expansion was 3h earlier than anticipation, validating that MCO has the potential for investigating ovarian toxic agents in vitro. We provide a novel approach for building an in vitro ovarian model to recapitulate ovarian functions and test chemical toxicity, suggesting it has the potential for clinical research in the future.
Collapse
Affiliation(s)
- Min Ye
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yiran Shan
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Bingchuan Lu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Hao Luo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Binhan Li
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Zixuan Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yuzhi Guo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Liliang Ouyang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
- Corresponding author. Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
- Corresponding author. Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
4
|
Maji S, Lee H. Engineering Hydrogels for the Development of Three-Dimensional In Vitro Models. Int J Mol Sci 2022; 23:2662. [PMID: 35269803 PMCID: PMC8910155 DOI: 10.3390/ijms23052662] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 02/06/2023] Open
Abstract
The superiority of in vitro 3D cultures over conventional 2D cell cultures is well recognized by the scientific community for its relevance in mimicking the native tissue architecture and functionality. The recent paradigm shift in the field of tissue engineering toward the development of 3D in vitro models can be realized with its myriad of applications, including drug screening, developing alternative diagnostics, and regenerative medicine. Hydrogels are considered the most suitable biomaterial for developing an in vitro model owing to their similarity in features to the extracellular microenvironment of native tissue. In this review article, recent progress in the use of hydrogel-based biomaterial for the development of 3D in vitro biomimetic tissue models is highlighted. Discussions of hydrogel sources and the latest hybrid system with different combinations of biopolymers are also presented. The hydrogel crosslinking mechanism and design consideration are summarized, followed by different types of available hydrogel module systems along with recent microfabrication technologies. We also present the latest developments in engineering hydrogel-based 3D in vitro models targeting specific tissues. Finally, we discuss the challenges surrounding current in vitro platforms and 3D models in the light of future perspectives for an improved biomimetic in vitro organ system.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
- Department of Smart Health Science and Technology, Kangwon National University (KNU), Chuncheon 24341, Korea
| |
Collapse
|
5
|
Hu X, Xia Z, Cai K. Recent advances of 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B 2022; 10:1486-1507. [DOI: 10.1039/d1tb02537f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly recognized as resources for disease treatments and regenerative medicine. Meanwhile, the unique chemical and physical properties of hydrogels provide innate advantages to achieve...
Collapse
|
6
|
Genovese P, Patel A, Ziemkiewicz N, Paoli A, Bruns J, Case N, Zustiak SP, Garg K. Co-delivery of fibrin-laminin hydrogel with mesenchymal stem cell spheroids supports skeletal muscle regeneration following trauma. J Tissue Eng Regen Med 2021; 15:1131-1143. [PMID: 34551191 DOI: 10.1002/term.3243] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/09/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022]
Abstract
Volumetric muscle loss (VML) is traumatic or surgical loss of skeletal muscle with resultant functional impairment. Skeletal muscle's innate capacity for regeneration is lost with VML due to a critical loss of stem cells, extracellular matrix, and neuromuscular junctions. Consequences of VML include permanent disability or delayed amputations of the affected limb. Currently, a successful clinical therapy has not been identified. Mesenchymal stem cells (MSCs) possess regenerative and immunomodulatory properties and their three-dimensional aggregation can further enhance therapeutic efficacy. In this study, MSC aggregation into spheroids was optimized in vitro based on cellular viability, spheroid size, and trophic factor secretion. The regenerative potential of the optimized MSC spheroid therapy was then investigated in a murine model of VML injury. Experimental groups included an untreated VML injury control, intramuscular injection of MSC spheroids, and MSC spheroids encapsulated in a fibrin-laminin hydrogel. Compared to the untreated VML group, the spheroid encapsulating hydrogel group enhanced myogenic marker (i.e., MyoD and myogenin) protein expression, improved muscle mass, increased presence of centrally nucleated myofibers as well as small fibers (<500 μm2 ), modulated pro- and anti-inflammatory macrophage marker expression (i.e., iNOS and Arginase), and increased the presence of CD146+ pericytes and CD31+ endothelial cells in the VML injured muscles. Future studies will evaluate the extent of functional recovery with the spheroid encapsulating hydrogel therapy.
Collapse
Affiliation(s)
- Peter Genovese
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Anjali Patel
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Natalia Ziemkiewicz
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Allison Paoli
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Joseph Bruns
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Natasha Case
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Silviya P Zustiak
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| | - Koyal Garg
- Program of Biomedical Engineering, School of Engineering, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
7
|
Caldwell AS, Aguado BA, Anseth KS. Designing Microgels for Cell Culture and Controlled Assembly of Tissue Microenvironments. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1907670. [PMID: 33841061 PMCID: PMC8026140 DOI: 10.1002/adfm.201907670] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 05/04/2023]
Abstract
Micron-sized hydrogels, termed microgels, are emerging as multifunctional platforms that can recapitulate tissue heterogeneity in engineered cell microenvironments. The microgels can function as either individual cell culture units or can be assembled into larger scaffolds. In this manner, individual microgels can be customized for single or multi-cell co-culture applications, or heterogeneous populations can be used as building blocks to create microporous assembled scaffolds that more closely mimic tissue heterogeneities. The inherent versatility of these materials allows user-defined control of the microenvironments, from the order of singly encapsulated cells to entire three-dimensional cell scaffolds. These hydrogel scaffolds are promising for moving towards personalized medicine approaches and recapitulating the multifaceted microenvironments that exist in vivo.
Collapse
Affiliation(s)
- Alexander S. Caldwell
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| | - Brian A. Aguado
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| |
Collapse
|
8
|
Wang J, Yu Y, Guo J, Lu W, Wei Q, Zhao Y. The Construction and Application of Three-Dimensional Biomaterials. ACTA ACUST UNITED AC 2020; 4:e1900238. [PMID: 32293130 DOI: 10.1002/adbi.201900238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/26/2019] [Indexed: 12/14/2022]
Abstract
Biomaterials have been widely explored and applied in many areas, especially in the field of tissue engineering. The interface of biomaterials and cells has been deeply investigated. However, it has been demonstrated that conventional 2D biomaterials fail to maintain the 3D structures and phenotypes of cells, which is the result of their limited ability to mimic the latter's complex extracellular matrix. To overcome this challenge, cell cultivation dependent on 3D biomaterials has emerged as an alternative strategy to make the recovery of 3D structures and functions of cells possible. Thus, with the thriving development of 3D cell culture in tissue engineering, a holistic review of the construction and application of 3D biomaterials is desired. Here, recent developments in 3D biomaterials for tissue engineering are reviewed. An overview of various approaches to construct 3D biomaterials, such as electro-jetting/-spinning, micro-molding, microfluidics, and 3D bio-printing, is first presented. Their typical applications in constructing cell sheets, vascular structures, cell spheroids, and macroscopic cellular constructs are described as well. Following these two sections, the current status and challenges are analyzed, as well as the future outlook of 3D biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Jie Wang
- College of Engineering, Nanjing Agricultural University, Nanjing, 210031, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yunru Yu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jiahui Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Wei Lu
- College of Engineering, Nanjing Agricultural University, Nanjing, 210031, China
| | - Qiong Wei
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
9
|
Heo DN, Hospodiuk M, Ozbolat IT. Synergistic interplay between human MSCs and HUVECs in 3D spheroids laden in collagen/fibrin hydrogels for bone tissue engineering. Acta Biomater 2019; 95:348-356. [PMID: 30831326 DOI: 10.1016/j.actbio.2019.02.046] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/17/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023]
Abstract
Stem cell encapsulation in hydrogels has been widely employed in tissue engineering, regenerative medicine, organ-on-a-chip devices and gene delivery; however, fabrication of native-like bone tissue using such a strategy has been a challenge, particularly in vitro, due to the limited cell loading densities resulting in weaker cell-cell interactions and lesser extra-cellular matrix deposition. In particular, scalable bone tissue constructs require vascular network to provide enough oxygen and nutrient supplies to encapsulated cells. To enhance stem cell function and generate pre-vascularized network, we here employed collagen/fibrin hydrogel as an encapsulation matrix for the incorporation of human mesenchymal stem cell/human umbilical vein endothelial cell (MSC/HUVEC) spheroids, and investigated their cellular behavior (including cell viability, morphology, proliferation, and gene expression profile) and compared to that of cell suspension- or MSC spheroids-laden hydrogels. MSC/HUVEC spheroids encapsulated in collagen/fibrin hydrogel showed better cell spreading and proliferation, and up-regulated osteogenic differentiation, and demonstrated pre-vascular network formation. Overall, MSC/HUVEC spheroids-laden hydrogels provided a highly suitable 3D microenvironment for bone tissue formation, which can be utilized in various applications, such as but not limited to tissue engineering, disease modeling and drug screening. STATEMENT OF SIGNIFICANCE: Stem cell encapsulation in hydrogels has been widely used in various areas such as tissue engineering, regenerative medicine, organ-on-a-chip devices and gene delivery; however, fabrication of native-like bone tissue using such an approach has been a challenge, particularly in vitro, due to the limited cell loading densities resulting in weaker cell-cell interactions and lesser extra-cellular matrix deposition. Here in this work, we have encapsulated spheroids of human mesenchymal stems cells (MSCs) in collagen/fibrin hydrogel and evaluated their viability, proliferation, osteogenic differentiation, and bone formation potential in vitro with respect to cell suspension-laden hydrogel samples. We have further incorporated human umbilical vein endothelial cells (HUVECs) into MSC spheroids and demonstrated that the presence of HUVECs in 3D spheroid culture in collagen/fibrin gel induced the formation of pre-vascular network, improved cell viability and proliferation, enhanced the osteogenic differentiation of spheroids, and increased their bone mineral deposition. In sum, MSC/HUVEC spheroids laden hydrogels provided a highly suitable 3D microenvironment for bone tissue formation, which can be utilized in various applications, such as but not limited to tissue engineering and regenerative medicine, disease modeling and drug screening.
Collapse
|
10
|
Leferink A, Tibbe M, Bossink E, de Heus L, van Vossen H, van den Berg A, Moroni L, Truckenmüller R. Shape-defined solid micro-objects from poly(d,l-lactic acid) as cell-supportive counterparts in bottom-up tissue engineering. Mater Today Bio 2019; 4:100025. [PMID: 32159154 PMCID: PMC7061620 DOI: 10.1016/j.mtbio.2019.100025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 01/01/2023] Open
Abstract
In bottom-up tissue engineering, small modular units of cells and biomaterials are assembled toward larger and more complex ones. In conjunction with a new implementation of this approach, a novel method to fabricate microscale objects from biopolymers by thermal imprinting on water-soluble sacrificial layers is presented. By this means, geometrically well-defined objects could be obtained without involving toxic agents in the form of photoinitiators. The micro-objects were used as cell-adhesive substrates and cell spacers in engineered tissues created by cell-guided assembly of the objects. Such constructs can be applied both for in vitro studies and clinical treatments. Clinically relevantly sized aggregates comprised of cells and micro-objects retained their viability up to 2 weeks of culture. The aggregation behavior of cells and objects showed to depend on the type and number of cells applied. To demonstrate the micro-objects' potential for engineering vascularized tissues, small aggregates of human bone marrow stromal cells (hMSCs) and micro-objects were coated with a layer of human umbilical vein endothelial cells (HUVECs) and fused into larger tissue constructs, resulting in HUVEC-rich regions at the aggregates' interfaces. This three-dimensional network-type spatial cellular organization could foster the establishment of (premature) vascular structures as a vital prerequisite of, for example, bottom-up-engineered bone-like tissue.
Collapse
Affiliation(s)
- A.M. Leferink
- Applied Stem Cell Technologies Group, TechMed Centre, University of Twente, 7500 AE, Enschede, the Netherlands
- BIOS/Lab on a Chip Group, TechMed Centre and MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, the Netherlands
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - M.P. Tibbe
- BIOS/Lab on a Chip Group, TechMed Centre and MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, the Netherlands
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - E.G.B.M. Bossink
- BIOS/Lab on a Chip Group, TechMed Centre and MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, the Netherlands
| | - L.E. de Heus
- Applied Stem Cell Technologies Group, TechMed Centre, University of Twente, 7500 AE, Enschede, the Netherlands
- BIOS/Lab on a Chip Group, TechMed Centre and MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, the Netherlands
| | - H. van Vossen
- MESA+ NanoLab, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, the Netherlands
| | - A. van den Berg
- BIOS/Lab on a Chip Group, TechMed Centre and MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, the Netherlands
| | - L. Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - R.K. Truckenmüller
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
11
|
Wechsler ME, Stephenson RE, Murphy AC, Oldenkamp HF, Singh A, Peppas NA. Engineered microscale hydrogels for drug delivery, cell therapy, and sequencing. Biomed Microdevices 2019; 21:31. [PMID: 30904963 DOI: 10.1007/s10544-019-0358-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Engineered microscale hydrogels have emerged as promising therapeutic approaches for the treatment of various diseases. These microgels find wide application in the biomedical field because of the ease of injectability, controlled release of therapeutics, flexible means of synthesis, associated tunability, and can be engineered as stimuli-responsive. While bulk hydrogels of several length-scale dimensions have been used for over two decades in drug delivery applications, their use as microscale carriers of drug and cell-based therapies is relatively new. Herein, we critically summarize the fundamentals of hydrogels based on their equilibrium and dynamics of their molecular structure, as well as solute diffusion as it relates to drug delivery. In addition, examples of common microgel synthesis techniques are provided. The ability to tune microscale hydrogels to obtain controlled release of therapeutics is discussed, along with microgel considerations for cell encapsulation as it relates to the development of cell-based therapies. We conclude with an outlook on the use of microgels for cell sequencing, and the convergence of the use of microscale hydrogels for drug delivery, cell therapy, and cell sequencing based systems.
Collapse
Affiliation(s)
- Marissa E Wechsler
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Regan E Stephenson
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Andrew C Murphy
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Heidi F Oldenkamp
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Ankur Singh
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA.
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.
- Department of Surgery and Perioperative Care, and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
12
|
Kim H, Bae C, Kook YM, Koh WG, Lee K, Park MH. Mesenchymal stem cell 3D encapsulation technologies for biomimetic microenvironment in tissue regeneration. Stem Cell Res Ther 2019; 10:51. [PMID: 30732645 PMCID: PMC6367797 DOI: 10.1186/s13287-018-1130-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell (MSC) encapsulation technique has long been emerged in tissue engineering as it plays an important role in implantation of stem cells to regenerate a damaged tissue. MSC encapsulation provides a mimic of a three-dimensional (3D) in vivo environment to maintain cell viability and to induce the stem cell differentiation which regulates MSC fate into multi-lineages. Moreover, the 3D matrix surrounding MSCs protects them from the human innate immune system and allows the diffusion of biomolecules such as oxygen, cytokines, and growth factors. Therefore, many technologies are being developed to create MSC encapsulation platforms with diverse materials, shapes, and sizes. The conditions of the platform are determined by the targeted tissue and translation method. This review introduces several details of MSC encapsulation technologies such as micromolding, electrostatic droplet extrusion, microfluidics, and bioprinting and their application for tissue regeneration. Lastly, some of the challenges and future direction of MSC encapsulation technologies as a cell therapy-based tissue regeneration method will be discussed.
Collapse
Affiliation(s)
- Hyerim Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Chaewon Bae
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Yun-Min Kook
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Kangwon Lee
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea. .,Advanced Institutes of Convergence Technology, Suwon, Republic of Korea.
| | - Min Hee Park
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea. .,Center for Convergence Bioceramic Materials, Korea Institute of Ceramic Engineering and Technology, Cheongju, Republic of Korea.
| |
Collapse
|
13
|
Huang PJ, Qu J, Saha P, Muliana A, Kameoka J. Microencapsulation of beta cells in collagen micro-disks via circular pneumatically actuated soft micro-mold (cPASMO) device. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aae55e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
14
|
Correia CR, Reis RL, Mano JF. Design Principles and Multifunctionality in Cell Encapsulation Systems for Tissue Regeneration. Adv Healthc Mater 2018; 7:e1701444. [PMID: 30102458 DOI: 10.1002/adhm.201701444] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Cell encapsulation systems are being increasingly applied as multifunctional strategies to regenerate tissues. Lessons afforded with encapsulation systems aiming to treat endocrine diseases seem to be highly valuable for the tissue engineering and regenerative medicine (TERM) systems of today, in which tissue regeneration and biomaterial integration are key components. Innumerous multifunctional systems for cell compartmentalization are being proposed to meet the specific needs required in the TERM field. Herein is reviewed the variable geometries proposed to produce cell encapsulation strategies toward tissue regeneration, including spherical and fiber-shaped systems, and other complex shapes and arrangements that better mimic the highly hierarchical organization of native tissues. The application of such principles in the TERM field brings new possibilities for the development of highly complex systems, which holds tremendous promise for tissue regeneration. The complex systems aim to recreate adequate environmental signals found in native tissue (in particular during the regenerative process) to control the cellular outcome, and conferring multifunctional properties, namely the incorporation of bioactive molecules and the ability to create smart and adaptative systems in response to different stimuli. The new multifunctional properties of such systems that are being employed to fulfill the requirements of the TERM field are also discussed.
Collapse
Affiliation(s)
- Clara R. Correia
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - João F. Mano
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| |
Collapse
|
15
|
|
16
|
Huang PJ, Chou CK, Chen CT, Yamaguchi H, Qu J, Muliana A, Hung MC, Kameoka J. Pneumatically Actuated Soft Micromold Device for Fabricating Collagen and Matrigel Microparticles. Soft Robot 2017; 4:390-399. [DOI: 10.1089/soro.2016.0073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Po-Jung Huang
- Department of Material Science and Engineering, Texas A&M University, College Station, Texas
| | - Chao-Kai Chou
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chun-Te Chen
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hirohito Yamaguchi
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jian Qu
- Department of Mechanical Engineering, Texas A&M University, College Station, Texas
| | - Anastasia Muliana
- Department of Mechanical Engineering, Texas A&M University, College Station, Texas
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Jun Kameoka
- Department of Material Science and Engineering, Texas A&M University, College Station, Texas
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas
- School of Medicine, The Jikei University, Tokyo, Japan
| |
Collapse
|
17
|
Affiliation(s)
- Suwan N. Jayasinghe
- BioPhysics Group, UCL Centre for Stem Cells and Regenerative Medicine; UCL Department of Mechanical Engineering and UCL Institute of Healthcare Engineering; University College London; Torrington Place London WC1E 7JE United Kingdom
| |
Collapse
|
18
|
Li F, Truong VX, Thissen H, Frith JE, Forsythe JS. Microfluidic Encapsulation of Human Mesenchymal Stem Cells for Articular Cartilage Tissue Regeneration. ACS APPLIED MATERIALS & INTERFACES 2017; 9:8589-8601. [PMID: 28225583 DOI: 10.1021/acsami.7b00728] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Stem cell injections for the treatment of articular cartilage damage are a promising approach to achieve tissue regeneration. However, this method is encumbered by high cell apoptosis rates, low retention in the cartilage lesion, and inefficient chondrogenesis. Here, we have used a facile, very low cost-based microfluidic technique to create visible light-cured microgels composed of gelatin norbornene (GelNB) and a poly(ethylene glycol) (PEG) cross-linker. In addition, we have demonstrated that the process enables the rapid in situ microencapsulation of human bone marrow-derived mesenchymal stem cells (hBMSCs) under biocompatible microfluidic-processing conditions for long-term maintenance. The hBMSCs exhibited an unusually high degree of chondrogenesis in the GelNB microgels with chondro-inductive media, specifically toward the hyaline cartilage structure, with significant upregulation in type II collagen expression compared to the bulk hydrogel and "gold standard" pellet culture. Overall, we have demonstrated that these protein-based microgels can be engineered as promising therapeutic candidates for articular cartilage regeneration, with additional potential to be used in a variety of other applications in regenerative medicine.
Collapse
Affiliation(s)
- Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University , Wellington Road, Clayton, VIC 3800, Australia
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - Vinh X Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University , Wellington Road, Clayton, VIC 3800, Australia
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University , Wellington Road, Clayton, VIC 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University , Wellington Road, Clayton, VIC 3800, Australia
| |
Collapse
|
19
|
Cui X, Hartanto Y, Zhang H. Advances in multicellular spheroids formation. J R Soc Interface 2017; 14:20160877. [PMID: 28202590 PMCID: PMC5332573 DOI: 10.1098/rsif.2016.0877] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/11/2017] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional multicellular spheroids (MCSs) have a complex architectural structure, dynamic cell-cell/cell-matrix interactions and bio-mimicking in vivo microenvironment. As a fundamental building block for tissue reconstruction, MCSs have emerged as a powerful tool to narrow down the gap between the in vitro and in vivo model. In this review paper, we discussed the structure and biology of MCSs and detailed fabricating methods. Among these methods, the approach in microfluidics with hydrogel support for MCS formation is promising because it allows essential cell-cell/cell-matrix interactions in a confined space.
Collapse
Affiliation(s)
- X Cui
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Y Hartanto
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - H Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
20
|
Samarasinghe SAPL, Shao Y, Huang PJ, Pishko M, Chu KH, Kameoka J. Fabrication of Bacteria Environment Cubes with Dry Lift-Off Fabrication Process for Enhanced Nitrification. PLoS One 2016; 11:e0165839. [PMID: 27812154 PMCID: PMC5094588 DOI: 10.1371/journal.pone.0165839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/18/2016] [Indexed: 12/11/2022] Open
Abstract
We have developed a 3D dry lift-off process to localize multiple types of nitrifying bacteria in polyethylene glycol diacrylate (PEGDA) cubes for enhanced nitrification, a two-step biological process that converts ammonium to nitrite and then to nitrate. Ammonia-oxidizing bacteria (AOB) is responsible for converting ammonia into nitrite, and nitrite-oxidizing bacteria (NOB) is responsible for converting nitrite to nitrate. Successful nitrification is often challenging to accomplish, in part because AOB and NOB are slow growers and highly susceptible to many organic and inorganic chemicals in wastewater. Most importantly, the transportation of chemicals among scattered bacteria is extremely inefficient and can be problematic. For example, nitrite, produced from ammonia oxidation, is toxic to AOB and can lead to the failure of nitrification. To address these challenges, we closely localize AOB and NOB in PEGDA cubes as microenvironment modules to promote synergetic interactions. The AOB is first localized in the vicinity of the surface of the PEGDA cubes that enable AOB to efficiently uptake ammonia from a liquid medium and convert it into nitrite. The produced nitrite is then efficiently transported to the NOB localized at the center of the PEGDA particle and converted into non-toxic nitrate. Additionally, the nanoscale PEGDA fibrous structures offer a protective environment for these strains, defending them from sudden toxic chemical shocks and immobilize in cubes. This engineered microenvironment cube significantly enhances nitrification and improves the overall ammonia removal rate per single AOB cell. This approach—encapsulation of multiple strains at close range in cube in order to control their interactions—not only offers a new strategy for enhancing nitrification, but also can be adapted to improve the production of fermentation products and biofuel, because microbial processes require synergetic reactions among multiple species.
Collapse
Affiliation(s)
- S. A. P. L. Samarasinghe
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Yiru Shao
- Zachry Department of Civil Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Po-Jung Huang
- Department of Material Science and Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Michael Pishko
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Kung-Hui Chu
- Zachry Department of Civil Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Jun Kameoka
- Department of Electrical & Computer Engineering, Texas A&M University, College Station, Texas, United States of America
- Department of Material Science and Engineering, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
21
|
Ma C, Tian C, Zhao L, Wang J. Pneumatic-aided micro-molding for flexible fabrication of homogeneous and heterogeneous cell-laden microgels. LAB ON A CHIP 2016; 16:2609-2617. [PMID: 27229899 DOI: 10.1039/c6lc00540c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Microgels are favorable for numerous applications such as drug delivery, biomaterials science and tissue engineering. Conventionally, photolithographic methods and micro-molding techniques are extensively exploited to prepare microgels; however, they are, respectively, limited to photocrosslinkable polymers and inadequate to generate serially patterned hydrogels due to the static nature of utilized molds. Herein, we proposed a simple and versatile approach, termed pneumatic-aided micro-molding (PAM), to flexibly fabricate microgels with precise control over multiple cell types and microarchitectures of hydrogels through strategically designed pneumatic microvalves. Using the PAM approach, different cells were encapsulated in various hydrogels that had well-defined geometries. Additionally, single/multiple micro-channeled cell-laden microgels were fabricated, of which the shape, number and arrangement could be finely tuned by varying microvalve configurations. Moreover, multi-compartmental microgels comprising composite hydrogel structures were engineered following a two-step PAM, which demonstrated the utility for biomimetically constructing a three-dimensional (3D) liver microtissue composed of a radially orchestrated network of hepatic cords and sinusoids. The resulting microtissue resembled the organizational complexity of the liver lobule and was applied for the evaluation of acetaminophen-induced hepatotoxicity. Collectively, the PAM strategy could be a useful and powerful tool in biomedical engineering, in vitro 3D cell culture, and fundamental biological studies.
Collapse
Affiliation(s)
- Chao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Chang Tian
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Lei Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Jinyi Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China. and College of Science, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
22
|
Abstract
Molecular diffusive membranes or materials are important for biological applications in microfluidic systems. Hydrogels are typical materials that offer several advantages, such as free diffusion for small molecules, biocompatibility with most cells, temperature sensitivity, relatively low cost, and ease of production. With the development of microfluidic applications, hydrogels can be integrated into microfluidic systems by soft lithography, flow-solid processes or UV cure methods. Due to their special properties, hydrogels are widely used as fluid control modules, biochemical reaction modules or biological application modules in different applications. Although hydrogels have been used in microfluidic systems for more than ten years, many hydrogels' properties and integrated techniques have not been carefully elaborated. Here, we systematically review the physical properties of hydrogels, general methods for gel-microfluidics integration and applications of this field. Advanced topics and the outlook of hydrogel fabrication and applications are also discussed. We hope this review can help researchers choose suitable methods for their applications using hydrogels.
Collapse
Affiliation(s)
- Xuanqi Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | | | | |
Collapse
|
23
|
Chan HF, Ma S, Leong KW. Can microfluidics address biomanufacturing challenges in drug/gene/cell therapies? Regen Biomater 2016; 3:87-98. [PMID: 27047674 PMCID: PMC4817324 DOI: 10.1093/rb/rbw009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 01/18/2016] [Indexed: 12/15/2022] Open
Abstract
Translation of any inventions into products requires manufacturing. Development of drug/gene/cell delivery systems will eventually face manufacturing challenges, which require the establishment of standardized processes to produce biologically-relevant products of high quality without incurring prohibitive cost. Microfluidicu technologies present many advantages to improve the quality of drug/gene/cell delivery systems. They also offer the benefits of automation. What remains unclear is whether they can meet the scale-up requirement. In this perspective, we discuss the advantages of microfluidic-assisted synthesis of nanoscale drug/gene delivery systems, formation of microscale drug/cell-encapsulated particles, generation of genetically engineered cells and fabrication of macroscale drug/cell-loaded micro-/nano-fibers. We also highlight the scale-up challenges one would face in adopting microfluidic technologies for the manufacturing of these therapeutic delivery systems.
Collapse
Affiliation(s)
- Hon Fai Chan
- Department of Biomedical Engineering, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Siying Ma
- Department of Biomedical Engineering, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
24
|
Green DW, Lee JS, Jung HS. Small-Scale Fabrication of Biomimetic Structures for Periodontal Regeneration. Front Physiol 2016; 7:6. [PMID: 26903872 PMCID: PMC4751709 DOI: 10.3389/fphys.2016.00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/08/2016] [Indexed: 11/18/2022] Open
Abstract
The periodontium is the supporting tissues for the tooth organ and is vulnerable to destruction, arising from overpopulating pathogenic bacteria and spirochaetes. The presence of microbes together with host responses can destroy large parts of the periodontium sometimes leading tooth loss. Permanent tissue replacements are made possible with tissue engineering techniques. However, existing periodontal biomaterials cannot promote proper tissue architectures, necessary tissue volumes within the periodontal pocket and a "water-tight" barrier, to become clinically acceptable. New kinds of small-scale engineered biomaterials, with increasing biological complexity are needed to guide proper biomimetic regeneration of periodontal tissues. So the ability to make compound structures with small modules, filled with tissue components, is a promising design strategy for simulating the anatomical complexity of the periodotium attachment complexes along the tooth root and the abutment with the tooth collar. Anatomical structures such as, intima, adventitia, and special compartments such as the epithelial cell rests of Malassez or a stellate reticulum niche need to be engineered from the start of regeneration to produce proper periodontium replacement. It is our contention that the positioning of tissue components at the origin is also necessary to promote self-organizing cell-cell connections, cell-matrix connections. This leads to accelerated, synchronized and well-formed tissue architectures and anatomies. This strategy is a highly effective preparation for tackling periodontitis, periodontium tissue resorption, and to ultimately prevent tooth loss. Furthermore, such biomimetic tissue replacements will tackle problems associated with dental implant support and perimimplantitis.
Collapse
Affiliation(s)
- David W. Green
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of DentistrySeoul, South Korea
- Oral Biosciences, Faculty of Dentistry, The University of Hong KongHong Kong, Hong Kong
| | - Jung-Seok Lee
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of DentistrySeoul, South Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of DentistrySeoul, South Korea
- Oral Biosciences, Faculty of Dentistry, The University of Hong KongHong Kong, Hong Kong
| |
Collapse
|
25
|
Morimoto Y, Hsiao AY, Takeuchi S. Point-, line-, and plane-shaped cellular constructs for 3D tissue assembly. Adv Drug Deliv Rev 2015; 95:29-39. [PMID: 26387835 DOI: 10.1016/j.addr.2015.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/18/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
Abstract
Microsized cellular constructs such as cellular aggregates and cell-laden hydrogel blocks are attractive cellular building blocks to reconstruct 3D macroscopic tissues with spatially ordered cells in bottom-up tissue engineering. In this regard, microfluidic techniques are remarkable methods to form microsized cellular constructs with high production rate and control of their shapes such as point, line, and plane. The fundamental shapes of the cellular constructs allow for the fabrication of larger arbitrary-shaped tissues by assembling them. This review introduces microfluidic formation methods of microsized cellular constructs and manipulation techniques to assemble them with control of their arrangements. Additionally, we show applications of the cellular constructs to biological studies and clinical treatments and discuss future trends as their potential applications.
Collapse
|
26
|
Liu J, Zheng H, Poh PSP, Machens HG, Schilling AF. Hydrogels for Engineering of Perfusable Vascular Networks. Int J Mol Sci 2015; 16:15997-6016. [PMID: 26184185 PMCID: PMC4519935 DOI: 10.3390/ijms160715997] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/09/2015] [Accepted: 07/07/2015] [Indexed: 02/03/2023] Open
Abstract
Hydrogels are commonly used biomaterials for tissue engineering. With their high-water content, good biocompatibility and biodegradability they resemble the natural extracellular environment and have been widely used as scaffolds for 3D cell culture and studies of cell biology. The possible size of such hydrogel constructs with embedded cells is limited by the cellular demand for oxygen and nutrients. For the fabrication of large and complex tissue constructs, vascular structures become necessary within the hydrogels to supply the encapsulated cells. In this review, we discuss the types of hydrogels that are currently used for the fabrication of constructs with embedded vascular networks, the key properties of hydrogels needed for this purpose and current techniques to engineer perfusable vascular structures into these hydrogels. We then discuss directions for future research aimed at engineering of vascularized tissue for implantation.
Collapse
Affiliation(s)
- Juan Liu
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany.
- Department of Hand Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Huaiyuan Zheng
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany.
- Department of Hand Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Patrina S P Poh
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany.
| | - Hans-Günther Machens
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany.
| | - Arndt F Schilling
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany.
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Science, D-80335 Munich, Germany.
| |
Collapse
|
27
|
Samorezov JE, Alsberg E. Spatial regulation of controlled bioactive factor delivery for bone tissue engineering. Adv Drug Deliv Rev 2015; 84:45-67. [PMID: 25445719 PMCID: PMC4428953 DOI: 10.1016/j.addr.2014.11.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 12/29/2022]
Abstract
Limitations of current treatment options for critical size bone defects create a significant clinical need for tissue engineered bone strategies. This review describes how control over the spatiotemporal delivery of growth factors, nucleic acids, and drugs and small molecules may aid in recapitulating signals present in bone development and healing, regenerating interfaces of bone with other connective tissues, and enhancing vascularization of tissue engineered bone. State-of-the-art technologies used to create spatially controlled patterns of bioactive factors on the surfaces of materials, to build up 3D materials with patterns of signal presentation within their bulk, and to pattern bioactive factor delivery after scaffold fabrication are presented, highlighting their applications in bone tissue engineering. As these techniques improve in areas such as spatial resolution and speed of patterning, they will continue to grow in value as model systems for understanding cell responses to spatially regulated bioactive factor signal presentation in vitro, and as strategies to investigate the capacity of the defined spatial arrangement of these signals to drive bone regeneration in vivo.
Collapse
Affiliation(s)
- Julia E Samorezov
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH, USA; National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
28
|
Kim J, Wu B, Niedzielski SM, Hill MT, Coleman RM, Ono A, Shikanov A. Characterizing natural hydrogel for reconstruction of three-dimensional lymphoid stromal network to model T-cell interactions. J Biomed Mater Res A 2015; 103:2701-10. [PMID: 25649205 DOI: 10.1002/jbm.a.35409] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/21/2015] [Accepted: 01/27/2015] [Indexed: 01/26/2023]
Abstract
Hydrogels have been used in regenerative medicine because they provide a three-dimensional environment similar to soft tissues, allow diffusion of nutrients, present critical biological signals, and degrade via endogenous enzymatic mechanisms. Herein, we developed in vitro system mimicking cell-cell and cell-matrix interactions in secondary lymphoid organs (SLOs). Existing in vitro culture systems cannot accurately represent the complex interactions happening between T-cells and stromal cells in immune response. To model T-cell interaction in SLOs in vitro, we encapsulated stromal cells in fibrin, collagen, or fibrin-collagen hydrogels and studied how different mechanical and biological properties affect stromal network formation. Overall, fibrin supplemented with aprotinin was superior to collagen and fibrin-collagen in terms of network formation and promotion of T-cell penetration. After 8 days of culture, stromal networks formed through branching and joining with other adjacent cell populations. T-cells added to the newly formed stromal networks migrated and attached to stromal cells, similar to the T-cell zones of the lymph nodes in vivo. Our results suggest that the constructed three-dimensional lymphoid stromal network can mimic the in vivo environment and allow the modeling of T-cell interaction in SLOs.
Collapse
Affiliation(s)
- Jiwon Kim
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan
| | - Biming Wu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | | | - Matthew T Hill
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Ariella Shikanov
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
29
|
Peterson AW, Caldwell DJ, Rioja AY, Rao RR, Putnam AJ, Stegemann JP. Vasculogenesis and Angiogenesis in Modular Collagen-Fibrin Microtissues. Biomater Sci 2014; 2:1497-1508. [PMID: 25177487 PMCID: PMC4145346 DOI: 10.1039/c4bm00141a] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The process of new blood vessel formation is critical in tissue development, remodeling and regeneration. Modular tissue engineering approaches have been developed to enable the bottom-up assembly of more complex tissues, including vascular networks. In this study, collagen-fibrin composite microbeads (100-300 μm in diameter) were fabricated using a water-in-oil emulsion technique. Human endothelial cells and human fibroblasts were embedded directly in the microbead matrix at the time of fabrication. Microbead populations were characterized and cultured for 14 days either as free-floating populations or embedded in a surrounding fibrin gel. The collagen-fibrin matrix efficiently entrapped cells and supported their viability and spreading. By 7 days in culture, endothelial cell networks were evident within microbeads, and these structures became more prominent by day 14. Fibroblasts co-localized with endothelial cells, suggesting a pericyte-like function, and laminin deposition indicated maturation of the vessel networks over time. Microbeads embedded in a fibrin gel immediately after fabrication showed the emergence of cells and the coalescence of vessel structures in the surrounding matrix by day 7. By day 14, inosculation of neighboring cords and prominent vessel structures were observed. Microbeads pre-cultured for 7 days prior to embedding in fibrin gave rise to vessel networks that emanated radially from the microbead by day 7, and developed into connected networks by day 14. Lumen formation in endothelial cell networks was confirmed using confocal sectioning. These data show that collagen-fibrin composite microbeads support vascular network formation. Microbeads embedded directly after fabrication emulated the process of vasculogenesis, while the branching and joining of vessels from pre-cultured microbeads resembled angiogenesis. This modular microtissue system has utility in studying the processes involved in new vessel formation, and may be developed into a therapy for the treatment of ischemic conditions.
Collapse
Affiliation(s)
- A W Peterson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - D J Caldwell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - A Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - R R Rao
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - A J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - J P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Walters BD, Stegemann JP. Strategies for directing the structure and function of three-dimensional collagen biomaterials across length scales. Acta Biomater 2014; 10:1488-501. [PMID: 24012608 PMCID: PMC3947739 DOI: 10.1016/j.actbio.2013.08.038] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/17/2013] [Accepted: 08/28/2013] [Indexed: 12/16/2022]
Abstract
Collagen type I is a widely used natural biomaterial that has found utility in a variety of biological and medical applications. Its well-characterized structure and role as an extracellular matrix protein make it a highly relevant material for controlling cell function and mimicking tissue properties. Collagen type I is abundant in a number of tissues, and can be isolated as a purified protein. This review focuses on hydrogel biomaterials made by reconstituting collagen type I from a solubilized form, with an emphasis on in vitro studies in which collagen structure can be controlled. The hierarchical structure of collagen from the nanoscale to the macroscale is described, with an emphasis on how structure is related to function across scales. Methods of reconstituting collagen into hydrogel materials are presented, including molding of macroscopic constructs, creation of microscale modules and electrospinning of nanoscale fibers. The modification of collagen biomaterials to achieve the desired structures and functions is also addressed, with particular emphasis on mechanical control of collagen structure, creation of collagen composite materials and crosslinking of collagenous matrices. Biomaterials scientists have made remarkable progress in rationally designing collagen-based biomaterials and in applying them both to the study of biology and for therapeutic benefit. This broad review illustrates recent examples of techniques used to control collagen structure and thereby to direct its biological and mechanical functions.
Collapse
Affiliation(s)
- B D Walters
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - J P Stegemann
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, MI 48109, USA.
| |
Collapse
|
31
|
Kang A, Park J, Ju J, Jeong GS, Lee SH. Cell encapsulation via microtechnologies. Biomaterials 2014; 35:2651-63. [PMID: 24439405 DOI: 10.1016/j.biomaterials.2013.12.073] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/20/2013] [Indexed: 01/01/2023]
Abstract
The encapsulation of living cells in a variety of soft polymers or hydrogels is important, particularly, for the rehabilitation of functional tissues capable of repairing or replacing damaged organs. Cellular encapsulation segregates cells from the surrounding tissue to protect the implanted cell from the recipient's immune system after transplantation. Diverse hydrogel membranes have been popularly used as encapsulating materials and permit the diffusion of gas, nutrients, wastes and therapeutic products smoothly. This review describes a variety of methods that have been developed to achieve cellular encapsulation using microscale platform. Microtechnologies have been adopted to precisely control the encapsulated cell number, size and shape of a cell-laden polymer structure. We provide a brief overview of recent microtechnology-based cell encapsulation methods, with a detailed description of the relevant processes. Finally, we discuss the current challenges and future directions likely to be taken by cell microencapsulation approaches toward tissue engineering and cell therapy applications.
Collapse
Affiliation(s)
- AhRan Kang
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea
| | - JiSoo Park
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea
| | - Jongil Ju
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-703, Republic of Korea
| | - Gi Seok Jeong
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-703, Republic of Korea
| | - Sang-Hoon Lee
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea; Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-703, Republic of Korea.
| |
Collapse
|
32
|
Schweicher J, Nyitray C, Desai TA. Membranes to achieve immunoprotection of transplanted islets. FRONT BIOSCI-LANDMRK 2014; 19:49-76. [PMID: 24389172 PMCID: PMC4230297 DOI: 10.2741/4195] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transplantation of islet or beta cells is seen as the cure for type 1 diabetes since it allows physiological regulation of blood glucose levels without requiring any compliance from the patients. In order to circumvent the use of immunosuppressive drugs (and their side effects), semipermeable membranes have been developed to encapsulate and immunoprotect transplanted cells. This review presents the historical developments of immunoisolation and provides an update on the current research in this field. A particular emphasis is laid on the fabrication, characterization and performance of membranes developed for immunoisolation applications.
Collapse
Affiliation(s)
- Julien Schweicher
- Therapeutic Micro and Nanotechnology Laboratory, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), 1700 4 Street, Box 2520, San Francisco, CA, 94158, USA
| | - Crystal Nyitray
- Therapeutic Micro and Nanotechnology Laboratory, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), 1700 4 Street, Box 2520, San Francisco, CA, 94158, USA
| | - Tejal A. Desai
- Therapeutic Micro and Nanotechnology Laboratory, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), 1700 4 Street, Box 2520, San Francisco, CA, 94158, USA
| |
Collapse
|
33
|
Guillotin B, Ali M, Ducom A, Catros S, Keriquel V, Souquet A, Remy M, Fricain JC, Guillemot F. Laser-Assisted Bioprinting for Tissue Engineering. Biofabrication 2013. [DOI: 10.1016/b978-1-4557-2852-7.00006-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Ciucurel EC, Chamberlain MD, Sefton MV. The Modular Approach. Biofabrication 2013. [DOI: 10.1016/b978-1-4557-2852-7.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Laser Assisted Bio-printing (LAB) of Cells and Bio-materials Based on Laser Induced Forward Transfer (LIFT). LASER TECHNOLOGY IN BIOMIMETICS 2013. [DOI: 10.1007/978-3-642-41341-4_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Morimoto Y, Takeuchi S. Three-dimensional cell culture based on microfluidic techniques to mimic living tissues. Biomater Sci 2012; 1:257-264. [PMID: 32481851 DOI: 10.1039/c2bm00117a] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This mini-review consists of microfluidic fabrication methods of cellular spheroids and cell-laden hydrogels, and their applications for tissue engineering. Using microfluidic devices, cellular spheroids and cell-laden hydrogels with controllable design are formed reproducibly. Owing to their size uniformity, they are used as building blocks for bottom-up tissue engineering to construct uniform and arbitrarily shaped tissues. Thus, cellular spheroids and cell-laden hydrogels based on microfluidic techniques are powerful tools to create tissues for human implantation and the treatment of diseases.
Collapse
Affiliation(s)
- Yuya Morimoto
- Center for International Research on Micronano Mechatronics (CIRMM), Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | | |
Collapse
|
37
|
Hu W, Ishii KS, Fan Q, Ohta AT. Hydrogel microrobots actuated by optically generated vapour bubbles. LAB ON A CHIP 2012; 12:3821-6. [PMID: 22899225 DOI: 10.1039/c2lc40483d] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
A novel hydrogel microrobot made of poly(ethylene glycol) diacrylate (PEGDA) is reported. This disk-shaped microrobot is optothermally actuated by laser-induced bubbles. A pulsed laser is used to smoothly actuate an 80-μm-diameter bubble microrobot at up to 320 μm s(-1). A single microrobot or a pair of microrobots working in cooperation were used to assemble 20-μm-diameter polystyrene beads into different patterns. The microrobots were also used to assemble patterns made of single yeast cells and cell-laden agarose microgels. The patterned yeast cells and cell-laden microgels were cultured, and the cells successfully multiplied.
Collapse
Affiliation(s)
- Wenqi Hu
- Dept. of Electrical Engineering, University of Hawaii at Manoa, Holmes Hall 483, 2540 Dole Street, Honolulu, USA
| | | | | | | |
Collapse
|
38
|
Janeczek Portalska K, Leferink A, Groen N, Fernandes H, Moroni L, van Blitterswijk C, de Boer J. Endothelial differentiation of mesenchymal stromal cells. PLoS One 2012; 7:e46842. [PMID: 23056481 PMCID: PMC3464214 DOI: 10.1371/journal.pone.0046842] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/06/2012] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stromal cells (hMSCs) are increasingly used in regenerative medicine for restoring worn-out or damaged tissue. Newly engineered tissues need to be properly vascularized and current candidates for in vitro tissue pre-vascularization are endothelial cells and endothelial progenitor cells. However, their use in therapy is hampered by their limited expansion capacity and lack of autologous sources. Our approach to engineering large grafts is to use hMSCs both as a source of cells for regeneration of targeted tissue and at the same time as the source of endothelial cells. Here we investigate how different stimuli influence endothelial differentiation of hMSCs. Although growth supplements together with shear force were not sufficient to differentiate hMSCs with respect to expression of endothelial markers such as CD31 and KDR, these conditions did prime the cells to differentiate into cells with an endothelial gene expression profile and morphology when seeded on Matrigel. In addition, we show that endothelial-like hMSCs are able to create a capillary network in 3D culture both in vitro and in vivo conditions. The expansion phase in the presence of growth supplements was crucial for the stability of the capillaries formed in vitro. To conclude, we established a robust protocol for endothelial differentiation of hMSCs, including an immortalized MSC line (iMSCs) which allows for reproducible in vitro analysis in further studies.
Collapse
Affiliation(s)
- Karolina Janeczek Portalska
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Anne Leferink
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Nathalie Groen
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Hugo Fernandes
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Lorenzo Moroni
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Clemens van Blitterswijk
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| | - Jan de Boer
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, The Netherlands
| |
Collapse
|
39
|
Coutinho DF, Ahari AF, Kachouie NN, Gomes ME, Neves NM, Reis RL, Khademhosseini A. An automated two-phase system for hydrogel microbead production. Biofabrication 2012; 4:035003. [PMID: 22914562 DOI: 10.1088/1758-5082/4/3/035003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymeric beads have been used for protection and delivery of bioactive materials, such as drugs and cells, for different biomedical applications. Here, we present a generic two-phase system for the production of polymeric microbeads of gellan gum or alginate, based on a combination of in situ polymerization and phase separation. Polymer droplets, dispensed using a syringe pump, formed polymeric microbeads while passing through a hydrophobic phase. These were then crosslinked, and thus stabilized, in a hydrophilic phase as they crossed through the hydrophobic-hydrophilic interface. The system can be adapted to different applications by replacing the bioactive material and the hydrophobic and/or the hydrophilic phases. The size of the microbeads was dependent on the system parameters, such as needle size and solution flow rate. The size and morphology of the microbeads produced by the proposed system were uniform, when parameters were kept constant. This system was successfully used for generating polymeric microbeads with encapsulated fluorescent beads, cell suspensions and cell aggregates proving its ability for generating bioactive carriers that can potentially be used for drug delivery and cell therapy.
Collapse
Affiliation(s)
- Daniela F Coutinho
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Taipas, 4806-909 Guimarães, Portugal
| | | | | | | | | | | | | |
Collapse
|
40
|
Matsui H, Takeuchi S, Osada T, Fujii T, Sakai Y. Enhanced bile canaliculi formation enabling direct recovery of biliary metabolites of hepatocytes in 3D collagen gel microcavities. LAB ON A CHIP 2012; 12:1857-1864. [PMID: 22441571 DOI: 10.1039/c2lc40046d] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Analysis of biliary metabolites is essential to predict pharmacokinetics and hepatotoxicity during drug development. In this paper, we present a hepatocyte culture configuration that enables the direct recovery of bile acid that accumulates in bile canaliculi by embedding the hepatocytes in a 3D micropatterned collagen gel substrate. We investigated the formation of bile canaliculi in hepatocytes embedded in circular microcavities of various sizes and made from collagen gel. Image analyses using fluorescently labeled bile acid revealed that the area of bile canaliculi in embedded hepatocytes in a microcavity of 60 or 80 μm in diameter was enlarged when compared with other sized microcavities and those of hepatocytes cultured using conventional hepatocyte sandwich cultures. We successfully recovered bile acid from the enlarged bile canaliculi of hepatocytes cultured in microcavities using a glass capillary and quantified the amount recovered. Using our approach, the direct recovery of biliary metabolites, using hepatocyte cultures with enhanced biliary excretion and geometrically enlarged bile canaliculi, may enable accurate screening of pharmacokinetics and drug-drug interactions against drug transporters.
Collapse
Affiliation(s)
- Hitoshi Matsui
- BEANS Laboratory, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | | | | | | | | |
Collapse
|
41
|
Fiddes LK, Luk VN, Au SH, Ng AHC, Luk V, Kumacheva E, Wheeler AR. Hydrogel discs for digital microfluidics. BIOMICROFLUIDICS 2012; 6:14112-1411211. [PMID: 22662096 PMCID: PMC3365348 DOI: 10.1063/1.3687381] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/27/2012] [Indexed: 05/10/2023]
Abstract
Hydrogels are networks of hydrophilic polymer chains that are swollen with water, and they are useful for a wide range of applications because they provide stable niches for immobilizing proteins and cells. We report here the marriage of hydrogels with digital microfluidic devices. Until recently, digital microfluidics, a fluid handling technique in which discrete droplets are manipulated electromechanically on the surface of an array of electrodes, has been used only for homogeneous systems involving liquid reagents. Here, we demonstrate for the first time that the cylindrical hydrogel discs can be incorporated into digital microfluidic systems and that these discs can be systematically addressed by droplets of reagents. Droplet movement is observed to be unimpeded by interaction with the gel discs, and gel discs remain stationary when droplets pass through them. Analyte transport into gel discs is observed to be identical to diffusion in cases in which droplets are incubated with gels passively, but transport is enhanced when droplets are continually actuated through the gels. The system is useful for generating integrated enzymatic microreactors and for three-dimensional cell culture. This paper demonstrates a new combination of techniques for lab-on-a-chip systems which we propose will be useful for a wide range of applications.
Collapse
|
42
|
Chen Z, Wang L, Stegemann JP. Phase-separated chitosan-fibrin microbeads for cell delivery. J Microencapsul 2012; 28:344-52. [PMID: 21736519 DOI: 10.3109/02652048.2011.569764] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Matrix-enhanced delivery of cells is a promising approach to improving current cell therapies. Our objective was to create cell-laden composite microbeads that combine the attractive features of the natural polymers chitosan and fibrin. Liquid polydimethylsiloxane was used to emulsify a chitosan-fibrinogen solution containing suspended human fibroblast cells, followed by initiation of thrombin-mediated polymerization of fibrin and thermal/pH-mediated gelation of chitosan. Chitosan/fibrin weight percent (wt%) ratios of 100/0, 75/25, 50/50 and 25/75 were investigated. Microbead diameters ranged from 275 ± 99 µm to 38 ± 10 µm using impeller speeds from 600 to 1400 rpm. Fibroblasts remained viable on day 1 post-fabrication in all matrices, but cell viability was markedly higher in high-fibrin microbeads by day 8 post-fabrication. Cell spreading and interaction with the extracellular matrix was also markedly increased in high-fibrin matrices. Such composite microbeads containing viable entrapped cells have potential for minimally invasive delivery of cells for a variety of tissue repair applications.
Collapse
Affiliation(s)
- Zhewei Chen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
43
|
Zheng H, Tian W, Yan H, Yue L, Zhang Y, Han F, Chen X, Li Y. Rotary culture promotes the proliferation of MCF-7 cells encapsulated in three-dimensional collagen-alginate hydrogels via activation of the ERK1/2-MAPK pathway. Biomed Mater 2012; 7:015003. [PMID: 22262729 DOI: 10.1088/1748-6041/7/1/015003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Rotary cell culture systems (RCCS) have been shown to be promising for promoting three-dimensional (3D) cell growth and assembly of cells into functional tissues. In this study, 3D tissue-like spheroids of MCF-7 cells were constructed by encapsulating the cells in the collagen-alginate hydrogel, and then cultured in a RCCS to investigate the proliferation of MCF-7 cells. The results from the MTT assay showed that the proliferation rate of MCF-7 cells cultured in the RCCS was higher than that of the static culture control group, and the results from the flow cytometry revealed that the cells in S and G2/M phase were significantly increased compared to the control group. The expression of cell proliferation antigen PCNA and cyclin D1 was also examined with the results further supporting the enhanced proliferation of MCF-7 cells by the RCCS. The results from indirect immunofluorescence revealed that the rotary culture altered neither the cytoskeleton distribution nor the assembly of mitotic spindle. By examination, it was also shown that the rotary culture induced the ERK1/2-MAPK pathway. Taken together, this study demonstrated that the rotary culture could promote the proliferation of MCF-7 cells by inducing the ERK1/2 pathway.
Collapse
Affiliation(s)
- Hongxia Zheng
- Department of Biological Science and Technology, Harbin Institute of Technology, Harbin, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Matalanis A, Jones OG, McClements DJ. Structured biopolymer-based delivery systems for encapsulation, protection, and release of lipophilic compounds. Food Hydrocoll 2011. [DOI: 10.1016/j.foodhyd.2011.04.014] [Citation(s) in RCA: 368] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
45
|
Komatsu H, Tsukiji S, Ikeda M, Hamachi I. Stiff, Multistimuli-Responsive Supramolecular Hydrogels as Unique Molds for 2D/3D Microarchitectures of Live Cells. Chem Asian J 2011; 6:2368-75. [DOI: 10.1002/asia.201100134] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Indexed: 11/06/2022]
|
46
|
Bai XP, Zheng HX, Fang R, Wang TR, Hou XL, Li Y, Chen XB, Tian WM. Fabrication of engineered heart tissue grafts from alginate/collagen barium composite microbeads. Biomed Mater 2011; 6:045002. [DOI: 10.1088/1748-6041/6/4/045002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Abstract
In vitro 3D culture is an important model for tissues in
vivo. Cells in different locations of 3D tissues are
physiologically different, because they are exposed to different concentrations
of oxygen, nutrients, and signaling molecules, and to other environmental
factors (temperature, mechanical stress, etc). The majority of high-throughput
assays based on 3D cultures, however, can only detect the
average behavior of cells in the whole 3D construct.
Isolation of cells from specific regions of 3D cultures is possible, but relies
on low-throughput techniques such as tissue sectioning and micromanipulation.
Based on a procedure reported previously (“cells-in-gels-in-paper”
or CiGiP), this paper describes a simple method for culture of arrays of thin
planar sections of tissues, either alone or stacked to create more complex 3D
tissue structures. This procedure starts with sheets of paper patterned with
hydrophobic regions that form 96 hydrophilic zones. Serial spotting of cells
suspended in extracellular matrix (ECM) gel onto the patterned paper creates an
array of 200 micron-thick slabs of ECM gel (supported mechanically by cellulose
fibers) containing cells. Stacking the sheets with zones aligned on top of one
another assembles 96 3D multilayer constructs. De-stacking the layers of the 3D
culture, by peeling apart the sheets of paper, “sections” all 96
cultures at once. It is, thus, simple to isolate 200-micron-thick
cell-containing slabs from each 3D culture in the 96-zone array. Because the 3D
cultures are assembled from multiple layers, the number of cells plated
initially in each layer determines the spatial distribution of cells in the
stacked 3D cultures. This capability made it possible to compare the growth of
3D tumor models of different spatial composition, and to examine the migration
of cells in these structures.
Collapse
|
48
|
Abstract
Recapitulating the elegant structures formed during development is an extreme synthetic and biological challenge. Great progress has been made in developing materials to support transplanted cells, yet the complexity of tissues is far beyond that found in even the most advanced scaffolds. Self-assembly is a motif used in development and a route for the production of complex materials. Self-assembly of peptides, proteins and other molecules at the nanoscale is promising, but in addition, intriguing ideas are emerging for self-assembly of micron-scale structures. In this brief review, very recent advances in the assembly of micron-scale cell aggregates and microgels will be described and discussed.
Collapse
|
49
|
Matsunaga YT, Morimoto Y, Takeuchi S. Molding cell beads for rapid construction of macroscopic 3D tissue architecture. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H90-4. [PMID: 21360782 DOI: 10.1002/adma.201004375] [Citation(s) in RCA: 204] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Indexed: 05/18/2023]
Affiliation(s)
- Yukiko T Matsunaga
- Institute of Industrial Science, The University of Tokyo, Komaba, Meguro-ku, Japan
| | | | | |
Collapse
|
50
|
Geckil H, Xu F, Zhang X, Moon S, Demirci U. Engineering hydrogels as extracellular matrix mimics. Nanomedicine (Lond) 2010; 5:469-84. [PMID: 20394538 DOI: 10.2217/nnm.10.12] [Citation(s) in RCA: 601] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular matrix (ECM) is a complex cellular environment consisting of proteins, proteoglycans, and other soluble molecules. ECM provides structural support to mammalian cells and a regulatory milieu with a variety of important cell functions, including assembling cells into various tissues and organs, regulating growth and cell-cell communication. Developing a tailored in vitro cell culture environment that mimics the intricate and organized nanoscale meshwork of native ECM is desirable. Recent studies have shown the potential of hydrogels to mimic native ECM. Such an engineered native-like ECM is more likely to provide cells with rational cues for diagnostic and therapeutic studies. The research for novel biomaterials has led to an extension of the scope and techniques used to fabricate biomimetic hydrogel scaffolds for tissue engineering and regenerative medicine applications. In this article, we detail the progress of the current state-of-the-art engineering methods to create cell-encapsulating hydrogel tissue constructs as well as their applications in in vitro models in biomedicine.
Collapse
Affiliation(s)
- Hikmet Geckil
- Health Sciences and Technology, Harvard-MIT Health Sciences and Technology, Bio-Acoustic-MEMS in Medicine (BAMM) Laboratory, 65 Landsdowne St., #267, 02139 Cambridge, MA, USA
| | | | | | | | | |
Collapse
|