1
|
Ashok D, Singh J, Howard HR, Cottam S, Waterhouse A, Bilek MMM. Interfacial engineering for biomolecule immobilisation in microfluidic devices. Biomaterials 2025; 316:123014. [PMID: 39708778 DOI: 10.1016/j.biomaterials.2024.123014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Microfluidic devices are used for various applications in biology and medicine. From on-chip modelling of human organs for drug screening and fast and straightforward point-of-care (POC) detection of diseases to sensitive biochemical analysis, these devices can be custom-engineered using low-cost techniques. The microchannel interface is essential for these applications, as it is the interface of immobilised biomolecules that promote cell capture, attachment and proliferation, sense analytes and metabolites or provide enzymatic reaction readouts. However, common microfluidic materials do not facilitate the stable immobilisation of biomolecules required for relevant applications, making interfacial engineering necessary to attach biomolecules to the microfluidic surfaces. Interfacial engineering is performed through various immobilisation mechanisms and surface treatment techniques, which suitably modify the surface properties like chemistry and energy to obtain robust biomolecule immobilisation and long-term storage stability suitable for the final application. In this review, we provide an overview of the status of interfacial engineering in microfluidic devices, covering applications, the role of biomolecules, their immobilisation pathways and the influence of microfluidic materials. We then propose treatment techniques to optimise performance for various biological and medical applications and highlight future areas of development.
Collapse
Affiliation(s)
- Deepu Ashok
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; Heart Research Institute, Newtown, NSW, 2042, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; Heart Research Institute, Newtown, NSW, 2042, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Henry Robert Howard
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sophie Cottam
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Marcela M M Bilek
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
2
|
Sun Y, Ikeuchi Y, Guo F, Hyun I, Ming GL, Fu J. Bioengineering innovations for neural organoids with enhanced fidelity and function. Cell Stem Cell 2025; 32:689-709. [PMID: 40315834 PMCID: PMC12052258 DOI: 10.1016/j.stem.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/19/2025] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Neural organoids have been utilized to recapitulate different aspects of the developing nervous system. While hailed as promising experimental tools for studying human neural development and neuropathology, current neural organoids do not fully recapitulate the anatomy or microcircuitry-level functionality of the developing brain, spinal cord, or peripheral nervous system. In this review, we discuss emerging bioengineering approaches that control morphogen signals and biophysical microenvironments, which have improved the efficiency, fidelity, and utility of neural organoids. Furthermore, advancements in bioengineered tools have facilitated more sophisticated analyses of neural organoid functions and applications, including improved neural-bioelectronic interfaces and organoid-based information processing. Emerging bioethical issues associated with advanced neural organoids are also discussed. Future opportunities of neural organoid research lie in enhancing their fidelity, maturity, and complexity and expanding their applications in a scalable manner.
Collapse
Affiliation(s)
- Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA.
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-8654, Japan
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47408, USA
| | - Insoo Hyun
- Center for Life Sciences and Public Learning, Museum of Science, Boston, MA 02114, USA; Center for Bioethics, Harvard Medical School, Boston, MA 02115, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Arora S, Singh S, Mittal A, Desai N, Khatri DK, Gugulothu D, Lather V, Pandita D, Vora LK. Spheroids in cancer research: Recent advances and opportunities. J Drug Deliv Sci Technol 2024; 100:106033. [DOI: 10.1016/j.jddst.2024.106033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
|
4
|
Pavon N, Sun Y, Pak C. Cell type specification and diversity in subpallial organoids. Front Genet 2024; 15:1440583. [PMID: 39391063 PMCID: PMC11465425 DOI: 10.3389/fgene.2024.1440583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Neural organoids have emerged as valuable tools for studying the developing brain, sparking enthusiasm and driving their adoption in disease modeling, drug screening, and investigating fetal neural development. The increasing popularity of neural organoids as models has led to a wide range of methodologies aimed at continuous improvement and refinement. Consequently, research groups often improve and reconfigure protocols to create region-specific organoids, resulting in diverse phenotypes, including variations in morphology, gene expression, and cell populations. While these improvements are exciting, routine adoptions of such modifications and protocols in the research laboratories are often challenging due to the reiterative empirical testing necessary to validate the cell types generated. To address this challenge, we systematically compare the similarities and differences that exist across published protocols that generates subpallial-specific organoids to date. In this review, we focus specifically on exploring the production of major GABAergic neuronal subtypes, especially Medium Spiny Neurons (MSNs) and Interneurons (INs), from multiple subpallial organoid protocols. Importantly, we look to evaluate the cell type diversity and the molecular pathways manipulated to generate them, thus broadening our understanding of the existing subpallial organoids as well as assessing the in vitro applicability of specific patterning factors. Lastly, we discuss the current challenges and outlook on the improved patterning of region-specific neural organoids. Given the critical roles MSN and IN dysfunction play in neurological disorders, comprehending the GABAergic neurons generated by neural organoids will undoubtedly facilitate clinical translation.
Collapse
Affiliation(s)
- Narciso Pavon
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Neuroscience and Behavior, University of Massachusetts Amherst, Amherst, MA, United States
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
5
|
López-León CF, Planet R, Soriano J. Preparation and Mechano-Functional Characterization of PEGylated Fibrin Hydrogels: Impact of Thrombin Concentration. Gels 2024; 10:116. [PMID: 38391447 PMCID: PMC10888336 DOI: 10.3390/gels10020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Three-dimensional (3D) neuronal cultures grown in hydrogels are promising platforms to design brain-like neuronal networks in vitro. However, the optimal properties of such cultures must be tuned to ensure a hydrogel matrix sufficiently porous to promote healthy development but also sufficiently rigid for structural support. Such an optimization is difficult since it implies the exploration of different hydrogel compositions and, at the same time, a functional analysis to validate neuronal culture viability. To advance in this quest, here we present a combination of a rheological protocol and a network-based functional analysis to investigate PEGylated fibrin hydrogel networks with gradually higher stiffness, achieved by increasing the concentration of thrombin. We observed that moderate thrombin concentrations of 10% and 25% in volume shaped healthy networks, although the functional traits depended on the hydrogel stiffness, which was much higher for the latter concentration. Thrombin concentrations of 65% or higher led to networks that did not survive. Our results illustrate the difficulties and limitations in preparing 3D neuronal networks, and stress the importance of combining a mechano-structural characterization of a biomaterial with a functional one.
Collapse
Affiliation(s)
- Clara F López-León
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelon, E-08028 Barcelona, Spain
| | - Ramon Planet
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelon, E-08028 Barcelona, Spain
| | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelon, E-08028 Barcelona, Spain
| |
Collapse
|
6
|
Pavon N, Diep K, Yang F, Sebastian R, Martinez-Martin B, Ranjan R, Sun Y, Pak C. Patterning ganglionic eminences in developing human brain organoids using a morphogen-gradient-inducing device. CELL REPORTS METHODS 2024; 4:100689. [PMID: 38228151 PMCID: PMC10831957 DOI: 10.1016/j.crmeth.2023.100689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/21/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
In early neurodevelopment, the central nervous system is established through the coordination of various neural organizers directing tissue patterning and cell differentiation. Better recapitulation of morphogen gradient production and signaling will be crucial for establishing improved developmental models of the brain in vitro. Here, we developed a method by assembling polydimethylsiloxane devices capable of generating a sustained chemical gradient to produce patterned brain organoids, which we termed morphogen-gradient-induced brain organoids (MIBOs). At 3.5 weeks, MIBOs replicated dorsal-ventral patterning observed in the ganglionic eminences (GE). Analysis of mature MIBOs through single-cell RNA sequencing revealed distinct dorsal GE-derived CALB2+ interneurons, medium spiny neurons, and medial GE-derived cell types. Finally, we demonstrate long-term culturing capabilities with MIBOs maintaining stable neural activity in cultures grown up to 5.5 months. MIBOs demonstrate a versatile approach for generating spatially patterned brain organoids for embryonic development and disease modeling.
Collapse
Affiliation(s)
- Narciso Pavon
- Graduate Program in Neuroscience and Behavior, UMass Amherst, Amherst, MA 01003, USA; Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Karmen Diep
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA 01003, USA
| | - Rebecca Sebastian
- Graduate Program in Neuroscience and Behavior, UMass Amherst, Amherst, MA 01003, USA; Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Beatriz Martinez-Martin
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA; Graduate Program in Molecular and Cellular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Ravi Ranjan
- Genomics Core, Institute of Applied Life Sciences, UMass Amherst, Amherst, MA 01003, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA 01003, USA.
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
7
|
Yang S, Xian Q, Liu Y, Zhang Z, Song Q, Gao Y, Wen W. A Silicon-Based PDMS-PEG Copolymer Microfluidic Chip for Real-Time Polymerase Chain Reaction Diagnosis. J Funct Biomater 2023; 14:jfb14040208. [PMID: 37103298 PMCID: PMC10143339 DOI: 10.3390/jfb14040208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/02/2023] [Accepted: 04/07/2023] [Indexed: 04/28/2023] Open
Abstract
Polydimethylsiloxane (PDMS) has been widely used to make lab-on-a-chip devices, such as reactors and sensors, for biological research. Real-time nucleic acid testing is one of the main applications of PDMS microfluidic chips due to their high biocompatibility and transparency. However, the inherent hydrophobicity and excessive gas permeability of PDMS hinder its applications in many fields. This study developed a silicon-based polydimethylsiloxane-polyethylene-glycol (PDMS-PEG) copolymer microfluidic chip, the PDMS-PEG copolymer silicon chip (PPc-Si chip), for biomolecular diagnosis. By adjusting the modifier formula for PDMS, the hydrophilic switch occurred within 15 s after contact with water, resulting in only a 0.8% reduction in transmittance after modification. In addition, we evaluated the transmittance at a wide range of wavelengths from 200 nm to 1000 nm to provide a reference for its optical property study and application in optical-related devices. The improved hydrophilicity was achieved by introducing a large number of hydroxyl groups, which also resulted in excellent bonding strength of PPc-Si chips. The bonding condition was easy to achieve and time-saving. Real-time PCR tests were successfully conducted with higher efficiency and lower non-specific absorption. This chip has a high potential for a wide range of applications in point-of-care tests (POCT) and rapid disease diagnosis.
Collapse
Affiliation(s)
- Siyu Yang
- Division of Emerging Interdisciplinary Areas, Interdisciplinary Program Office, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- Thrust of Advanced Materials, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen 518000, China
| | - Qingyue Xian
- Division of Emerging Interdisciplinary Areas, Interdisciplinary Program Office, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- Thrust of Advanced Materials, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
| | - Yiteng Liu
- Division of Emerging Interdisciplinary Areas, Interdisciplinary Program Office, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- Thrust of Advanced Materials, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
| | - Ziyi Zhang
- Division of Emerging Interdisciplinary Areas, Interdisciplinary Program Office, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- Thrust of Advanced Materials, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
| | - Qi Song
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Yibo Gao
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Weijia Wen
- Thrust of Advanced Materials, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen 518000, China
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
8
|
Sood A, Kumar A, Gupta VK, Kim CM, Han SS. Translational Nanomedicines Across Human Reproductive Organs Modeling on Microfluidic Chips: State-of-the-Art and Future Prospects. ACS Biomater Sci Eng 2023; 9:62-84. [PMID: 36541361 DOI: 10.1021/acsbiomaterials.2c01080] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Forecasting the consequence of nanoparticles (NPs) and therapeutically significant molecules before materializing for human clinical trials is a mainstay for drug delivery and screening processes. One of the noteworthy obstacles that has prevented the clinical translation of NP-based drug delivery systems and novel drugs is the lack of effective preclinical platforms. As a revolutionary technology, the organ-on-a-chip (OOC), a coalition of microfluidics and tissue engineering, has surfaced as an alternative to orthodox screening platforms. OOC technology recapitulates the structural and physiological features of human organs along with intercommunications between tissues on a chip. The current review discusses the concept of microfluidics and confers cutting-edge fabrication processes for chip designing. We also outlined the advantages of microfluidics in analyzing NPs in terms of characterization, transport, and degradation in biological systems. The review further elaborates the scope and research on translational nanomedicines in human reproductive organs (testis, placenta, uterus, and menstrual cycle) by taking the advantages offered by microfluidics and shedding light on their potential future implications. Finally, we accentuate the existing challenges for clinical translation and scale-up dynamics for microfluidics chips and emphasize its future perspectives.
Collapse
Affiliation(s)
- Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.,Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Vijai Kumar Gupta
- Biorefining and Advanced Materials Research Center, Scotland's Rural College, Edinburgh EH9 3JG, United Kingdom
| | - Chul Min Kim
- Department of Mechatronics Engineering, Gyeongsang National University, 33 Dongjin-ro, Jinju, Gyeongsangnam-do 52725, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.,Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| |
Collapse
|
9
|
Lee JW, Song KH. Fibrous hydrogels by electrospinning: Novel platforms for biomedical applications. J Tissue Eng 2023; 14:20417314231191881. [PMID: 37581121 PMCID: PMC10423451 DOI: 10.1177/20417314231191881] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023] Open
Abstract
Hydrogels, hydrophilic and biocompatible polymeric networks, have been used for numerous biomedical applications because they have exhibited abilities to mimic features of extracellular matrix (ECM). In particular, the hydrogels engineered with electrospinning techniques have shown great performances in biomedical applications. Electrospinning techniques are to generate polymeric micro/nanofibers that can mimic geometries of natural ECM by drawing micro/nanofibers from polymer precursors with electrical forces, followed by structural stabilization of them. By exploiting the electrospinning techniques, the fibrous hydrogels have been fabricated and utilized as 2D/3D cell culture platforms, implantable scaffolds, and wound dressings. In addition, some hydrogels that respond to external stimuli have been used to develop biosensors. For comprehensive understanding, this review covers electrospinning processes, hydrogel precursors used for electrospinning, characteristics of fibrous hydrogels and specific biomedical applications of electrospun fibrous hydrogels and highlight their potential to promote use in biomedical applications.
Collapse
Affiliation(s)
- Ji Woo Lee
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Kwang Hoon Song
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Republic of Korea
| |
Collapse
|
10
|
Iakovlev AP, Erofeev AS, Gorelkin PV. Novel Pumping Methods for Microfluidic Devices: A Comprehensive Review. BIOSENSORS 2022; 12:956. [PMID: 36354465 PMCID: PMC9688261 DOI: 10.3390/bios12110956] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 06/02/2023]
Abstract
This review is an account of methods that use various strategies to control microfluidic flow control with high accuracy. The reviewed systems are divided into two large groups based on the way they create flow: passive systems (non-mechanical systems) and active (mechanical) systems. Each group is presented by a number of device fabrications. We try to explain the main principles of operation, and we list advantages and disadvantages of the presented systems. Mechanical systems are considered in more detail, as they are currently an area of increased interest due to their unique precision flow control and "multitasking". These systems are often applied as mini-laboratories, working autonomously without any additional operations, provided by humans, which is very important under complicated conditions. We also reviewed the integration of autonomous microfluidic systems with a smartphone or single-board computer when all data are retrieved and processed without using a personal computer. In addition, we discuss future trends and possible solutions for further development of this area of technology.
Collapse
Affiliation(s)
| | | | - Petr V. Gorelkin
- Research Laboratory of Biophysics, National University of Science and Technology «MISiS», 119049 Moscow, Russia
| |
Collapse
|
11
|
Iyer NR, Ashton RS. Bioengineering the human spinal cord. Front Cell Dev Biol 2022; 10:942742. [PMID: 36092702 PMCID: PMC9458954 DOI: 10.3389/fcell.2022.942742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Three dimensional, self-assembled organoids that recapitulate key developmental and organizational events during embryogenesis have proven transformative for the study of human central nervous system (CNS) development, evolution, and disease pathology. Brain organoids have predominated the field, but human pluripotent stem cell (hPSC)-derived models of the spinal cord are on the rise. This has required piecing together the complex interactions between rostrocaudal patterning, which specifies axial diversity, and dorsoventral patterning, which establishes locomotor and somatosensory phenotypes. Here, we review how recent insights into neurodevelopmental biology have driven advancements in spinal organoid research, generating experimental models that have the potential to deepen our understanding of neural circuit development, central pattern generation (CPG), and neurodegenerative disease along the body axis. In addition, we discuss the application of bioengineering strategies to drive spinal tissue morphogenesis in vitro, current limitations, and future perspectives on these emerging model systems.
Collapse
Affiliation(s)
- Nisha R. Iyer
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wisconsin Institute for Discovery, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
| | - Randolph S. Ashton
- Wisconsin Institute for Discovery, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
| |
Collapse
|
12
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
13
|
Banik S, Uchil A, Kalsang T, Chakrabarty S, Ali MA, Srisungsitthisunti P, Mahato KK, Surdo S, Mazumder N. The revolution of PDMS microfluidics in cellular biology. Crit Rev Biotechnol 2022; 43:465-483. [PMID: 35410564 DOI: 10.1080/07388551.2022.2034733] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microfluidics is revolutionizing the way research on cellular biology has been traditionally conducted. The ability to control the cell physicochemical environment by adjusting flow conditions, while performing cellular analysis at single-cell resolution and high-throughput, has made microfluidics the ideal choice to replace traditional in vitro models. However, such a revolution only truly started with the advent of polydimethylsiloxane (PDMS) as a microfluidic structural material and soft-lithography as a rapid manufacturing technology. Indeed, before the "PDMS age," microfluidic technologies were: costly, time-consuming and, more importantly, accessible only to specialized laboratories and users. The simplicity of molding PDMS in various shapes along with its inherent properties (transparency, biocompatibility, and gas permeability) has spread the applications of innovative microfluidic devices to diverse and important biological fields and clinical studies. This review highlights how PDMS-based microfluidic systems are innovating pre-clinical biological research on cells and organs. These devices were able to cultivate different cell lines, enhance the sensitivity and diagnostic effectiveness of numerous cell-based assays by maintaining consistent chemical gradients, utilizing and detecting the smallest number of analytes while being high-throughput. This review will also assist in identifying the pitfalls in current PDMS-based microfluidic systems to facilitate breakthroughs and advancements in healthcare research.
Collapse
Affiliation(s)
- Soumyabrata Banik
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Ashwini Uchil
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Tenzin Kalsang
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Md Azahar Ali
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Pornsak Srisungsitthisunti
- Department of Production Engineering, Faculty of Engineering, King Mongkut's University of Technology North Bangkok, Bangkok, Thailand
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Salvatore Surdo
- Department of Nanophysics, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Nirmal Mazumder
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
14
|
Havins L, Capel A, Christie SD, Lewis MP, Roach P. Gradient biomimetic platforms for neurogenesis studies. J Neural Eng 2021; 19. [PMID: 34942614 DOI: 10.1088/1741-2552/ac4639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/23/2021] [Indexed: 01/09/2023]
Abstract
There is a need for the development of new cellular therapies for the treatment of many diseases, with the central nervous system (CNS) currently an area of specific focus. Due to the complexity and delicacy of its biology, there is currently a limited understanding of neurogenesis and consequently a lack of reliable test platforms, resulting in several CNS based diseases having no cure. The ability to differentiate pluripotent stem cells into specific neuronal sub-types may enable scalable manufacture for clinical therapies, with a focus also on the purity and quality of the cell population. This focus is targeted towards an urgent need for the diseases that currently have no cure, e.g. Parkinson's disease. Differentiation studies carried out using traditional 2D cell culture techniques are designed using biological signals and morphogens known to be important for neurogenesis in vivo. However, such studies are limited by their simplistic nature, including a general poor efficiency and reproducibility, high reagent costs and an inability to scale-up the process to a manufacture-wide design for clinical use. Biomimetic approaches to recapitulate a more in vivo-like environment are progressing rapidly within this field, with application of bio(chemical) gradients presented both as 2D surfaces and within a 3D volume. This review focusses on the development and application of these advanced extracellular environments particularly for the neural niche. We emphasise the progress that has been made specifically in the area of stem cell derived neuronal differentiation. Increasing developments in biomaterial approaches to manufacture stem cells will enable the improvement of differentiation protocols, enhancing the efficiency and repeatability of the process with a move towards up-scaling. Progress in this area brings these techniques closer to enabling the development of therapies for the clinic.
Collapse
Affiliation(s)
- Laurissa Havins
- Department of Chemistry, Loughborough University, Dept Chemistry, School of Science, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Andrew Capel
- Loughborough University, 2National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough, LE11 3TU, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Steven D Christie
- Department of Chemistry, Loughborough University, Dept Chemistry, School of Science, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Mark P Lewis
- Loughborough University School of Sport Exercise and Health Sciences, National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough, LE11 3TU, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Paul Roach
- Chemistry, Loughborough University, Dept Chemistry, School of Science, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| |
Collapse
|
15
|
Dogan E, Kisim A, Bati-Ayaz G, Kubicek GJ, Pesen-Okvur D, Miri AK. Cancer Stem Cells in Tumor Modeling: Challenges and Future Directions. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100017. [PMID: 34927168 PMCID: PMC8680587 DOI: 10.1002/anbr.202100017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Microfluidic tumors-on-chips models have revolutionized anticancer therapeutic research by creating an ideal microenvironment for cancer cells. The tumor microenvironment (TME) includes various cell types and cancer stem cells (CSCs), which are postulated to regulate the growth, invasion, and migratory behavior of tumor cells. In this review, the biological niches of the TME and cancer cell behavior focusing on the behavior of CSCs are summarized. Conventional cancer models such as three-dimensional cultures and organoid models are reviewed. Opportunities for the incorporation of CSCs with tumors-on-chips are then discussed for creating tumor invasion models. Such models will represent a paradigm shift in the cancer community by allowing oncologists and clinicians to predict better which cancer patients will benefit from chemotherapy treatments.
Collapse
Affiliation(s)
- Elvan Dogan
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028
| | - Asli Kisim
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey
| | - Gizem Bati-Ayaz
- Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Gregory J. Kubicek
- Department of Radiation Oncology, MD Anderson Cancer Center at Cooper, 2 Cooper Plaza, Camden, NJ 08103
| | - Devrim Pesen-Okvur
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey; Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028; School of Medical Engineering, Science, and Health, Rowan University, Camden, NJ 08103
| |
Collapse
|
16
|
Shakeri A, Khan S, Didar TF. Conventional and emerging strategies for the fabrication and functionalization of PDMS-based microfluidic devices. LAB ON A CHIP 2021; 21:3053-3075. [PMID: 34286800 DOI: 10.1039/d1lc00288k] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Microfluidics is an emerging and multidisciplinary field that is of great interest to manufacturers in medicine, biotechnology, and chemistry, as it provides unique tools for the development of point-of-care diagnostics, organs-on-chip systems, and biosensors. Polymeric microfluidics, unlike glass and silicon, offer several advantages such as low-cost mass manufacturing and a wide range of beneficial material properties, which make them the material of choice for commercial applications and high-throughput systems. Among polymers used for the fabrication of microfluidic devices, polydimethylsiloxane (PDMS) still remains the most widely used material in academia due to its advantageous properties, such as excellent transparency and biocompatibility. However, commercialization of PDMS has been a challenge mostly due to the high cost of the current fabrication strategies. Moreover, specific surface modification and functionalization steps are required to tailor the surface chemistry of PDMS channels (e.g. biomolecule immobilization, surface hydrophobicity and antifouling properties) with respect to the desired application. While significant research has been reported in the field of PDMS microfluidics, functionalization of PDMS surfaces remains a critical step in the fabrication process that is difficult to navigate. This review first offers a thorough illustration of existing fabrication methods for PDMS-based microfluidic devices, providing several recent advancements in this field with the aim of reducing the cost and time for mass production of these devices. Next, various conventional and emerging approaches for engineering the surface chemistry of PDMS are discussed in detail. We provide a wide range of functionalization techniques rendering PDMS microchannels highly biocompatible for physical or covalent immobilization of various biological entities while preventing non-specific interactions.
Collapse
Affiliation(s)
- Amid Shakeri
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada.
| | - Shadman Khan
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Tohid F Didar
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada.
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
17
|
Aghlmandi A, Nikshad A, Safaralizadeh R, Warkiani ME, Aghebati-Maleki L, Yousefi M. Microfluidics as efficient technology for the isolation and characterization of stem cells. EXCLI JOURNAL 2021; 20:426-443. [PMID: 33746671 PMCID: PMC7975637 DOI: 10.17179/excli2020-3028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/15/2021] [Indexed: 01/09/2023]
Abstract
The recent years have been passed with significant progressions in the utilization of microfluidic technologies for cellular investigations. The aim of microfluidics is to mimic small-scale body environment with features like optical transparency. Microfluidics can screen and monitor different cell types during culture and study cell function in response to stimuli in a fully controlled environment. No matter how the microfluidic environment is similar to in vivo environment, it is not possible to fully investigate stem cells behavior in response to stimuli during cell proliferation and differentiation. Researchers have used stem cells in different fields from fundamental researches to clinical applications. Many cells in the body possess particular functions, but stem cells do not have a specific task and can turn into almost any type of cells. Stem cells are undifferentiated cells with the ability of changing into specific cells that can be essential for the body. Researchers and physicians are interested in stem cells to use them in testing the function of the body's systems and solving their complications. This review discusses the recent advances in utilizing microfluidic techniques for the analysis of stem cells, and mentions the advantages and disadvantages of using microfluidic technology for stem cell research.
Collapse
Affiliation(s)
- Afsoon Aghlmandi
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Aylin Nikshad
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Majid Ebrahimi Warkiani
- The School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | | | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
18
|
Fannon OM, Bithell A, Whalley BJ, Delivopoulos E. A Fiber Alginate Co-culture Platform for the Differentiation of mESC and Modeling of the Neural Tube. Front Neurosci 2021; 14:524346. [PMID: 33510605 PMCID: PMC7835723 DOI: 10.3389/fnins.2020.524346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 12/04/2020] [Indexed: 12/28/2022] Open
Abstract
Alginate hydrogels are a commonly used substrate for in vitro 3D cell culture. These naturally derived biomaterials are highly tunable, biocompatible, and can be designed to mimic the elastic modulus of the adult brain at 1% w/v solution. Recent studies show that the molecular weight of the alginate can affect cell viability and differentiation. The relationship between the molecular weight, viscosity and ratio of G:M monomers of alginate hydrogels is complex, and the balance between these factors must be carefully considered when deciding on a suitable alginate hydrogel for stem cell research. This study investigates the formation of embryoid bodies (EB) from mouse embryonic stem cells, using low molecular weight (LMW) and high molecular weight (HMW) alginates. The cells are differentiated using a retinoic acid-based protocol, and the resulting aggregates are sectioned and stained for the presence of stem cells and the three germ layers (endoderm, mesoderm, and ectoderm). The results highlight that aggregates within LMW and HMW alginate are true EBs, as demonstrated by positive staining for markers of the three germ layers. Using tubular alginate scaffolds, formed with an adapted gradient maker protocol, we also propose a novel 3D platform for the patterned differentiation of mESCs, based on gradients of retinoic acid produced in situ by lateral motor column (LMC) motor neurons. The end product of our platform will be of great interest as it can be further developed into a powerful model of neural tube development.
Collapse
Affiliation(s)
- Orla M Fannon
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Angela Bithell
- School of Pharmacy, University of Reading, Reading, United Kingdom
| | | | | |
Collapse
|
19
|
Prunet A, Lefort S, Delanoë-Ayari H, Laperrousaz B, Simon G, Barentin C, Saci S, Argoul F, Guyot B, Rieu JP, Gobert S, Maguer-Satta V, Rivière C. A new agarose-based microsystem to investigate cell response to prolonged confinement. LAB ON A CHIP 2020; 20:4016-4030. [PMID: 32975276 DOI: 10.1039/d0lc00732c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Emerging evidence suggests the importance of mechanical stimuli in normal and pathological situations for the control of many critical cellular functions. While the effect of matrix stiffness has been and is still extensively studied, few studies have focused on the role of mechanical stresses. The main limitation of such analyses is the lack of standard in vitro assays enabling extended mechanical stimulation compatible with dynamic biological and biophysical cell characterization. We have developed an agarose-based microsystem, the soft cell confiner, which enables the precise control of confinement for single or mixed cell populations. The rigidity of the confiner matches physiological conditions and its porosity enables passive medium renewal. It is compatible with time-lapse microscopy, in situ immunostaining, and standard molecular analyses, and can be used with both adherent and non-adherent cell lines. Cell proliferation of various cell lines (hematopoietic cells, MCF10A epithelial breast cells and HS27A stromal cells) was followed for several days up to confluence using video-microscopy and further documented by Western blot and immunostaining. Interestingly, even though the nuclear projected area was much larger upon confinement, with many highly deformed nuclei (non-circular shape), cell viability, assessed by live and dead cell staining, was unaffected for up to 8 days in the confiner. However, there was a decrease in cell proliferation upon confinement for all cell lines tested. The soft cell confiner is thus a valuable tool to decipher the effects of long-term confinement and deformation on the biology of cell populations. This tool will be instrumental in deciphering the impact of nuclear and cytoskeletal mechanosensitivity in normal and pathological conditions involving highly confined situations, such as those reported upon aging with fibrosis or during cancer.
Collapse
Affiliation(s)
- A Prunet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5306, Institut Lumière Matière, F-69622, Villeurbanne, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Nie J, Fu J, He Y. Hydrogels: The Next Generation Body Materials for Microfluidic Chips? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003797. [PMID: 33103353 DOI: 10.1002/smll.202003797] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Indexed: 05/27/2023]
Abstract
The integration of microfluidics with biomedical research is confronted with considerable limitations due to its body materials. With high content of water, hydrogels own superior biocompatibility and degradability. Can hydrogels become another material choice for the construction of microfluidic chips, particularly biofluidics? The present review aims to systematically establish the concept of hydrogel-based microfluidic chips (HMCs) and address three main concerns: i) why choosing hydrogels? ii) how to fabricate HMCs?, and iii) in which fields to apply HMCs? It is envisioned that hydrogels may be used increasingly as substitute for traditional materials and gradually act as the body material for microfluidic chips. The modifications of conventional process are highlighted to overcome issues arising from the incompatibility between the construction methods and hydrogel materials. Specifically targeting at the "soft and wet" hydrogels, an efficient flowchart of "i) high resolution template printing; ii) damage-free demolding; iii) twice-crosslinking bonding" is proposed. Accordingly, a broader microfluidic chip concept is proposed in terms of form and function. Potential biomedical applications of HMCs are discussed. This review also highlights the challenges arising from the material replacement, as well as the future directions of the proposed concept. Finally, the authors' viewpoints and perspectives for this emerging field are discussed.
Collapse
Affiliation(s)
- Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
| |
Collapse
|
21
|
Xu J, Wang X, Li X, Yang G, Luo C. High-throughput cell migration assay under combinatorial chemical environments by a novel 24-well-plate based device. Biomed Microdevices 2020; 22:40. [PMID: 32474727 DOI: 10.1007/s10544-020-00491-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The quantitative studies of cell proliferation and migration under different chemical environments are important for both scientists and clinicians searching for new therapeutics. In this study, we developed a new device to pattern several types of cells in 24-well-plate and demonstrated its' application in cancer cell proliferation and migration assay. The new device combined 3D-printed-silica-part for multi cell types loading with PDMS-through-hole-layer-part for cell micro-patterning which was matched with commercial 24-well-plate. This 24-well-plate based device is flexible and feasible in many applications and can be used in one piece or multi pieces. Besides the application for two types of cells proliferation and migration assay in one chemical condition, as a demonstration, the migration behaviors of four types of cells under 24 types of EGF + bFGF combinatorial conditions were studied. We believed this device could be widely used in clinical searching for new anti-cancer therapeutics and other related studies.
Collapse
Affiliation(s)
- Jian Xu
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.,Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xudong Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, China
| | - Xiao Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, China.
| | - Chunxiong Luo
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China. .,Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
22
|
Tang SW, Tong WY, Pang SW, Voelcker NH, Lam YW. Deconstructing, Replicating, and Engineering Tissue Microenvironment for Stem Cell Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:540-554. [PMID: 32242476 DOI: 10.1089/ten.teb.2020.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.
Collapse
Affiliation(s)
- Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yin Tong
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Stella W Pang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong
| | - Nicolas H Voelcker
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
23
|
O'Grady BJ, Lippmann ES. Recent Advancements in Engineering Strategies for Manipulating Neural Stem Cell Behavior. ACTA ACUST UNITED AC 2020; 1:41-47. [PMID: 33748772 DOI: 10.1007/s43152-020-00003-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Purpose of Review Stem cells are exquisitely sensitive to biophysical and biochemical cues within the native microenvironment. This review focuses on emerging strategies to manipulate neural cell behavior using these influences in three-dimensional (3D) culture systems. Recent Findings Traditional systems for neural cell differentiation typically produce heterogeneous populations with limited diversity rather than the complex, organized tissue structures observed in vivo. Advancements in developing engineering tools to direct neural cell fates can enable new applications in basic research, disease modeling, and regenerative medicine. Summary This review article highlights engineering strategies that facilitate controlled presentation of biophysical and biochemical cues to guide differentiation and impart desired phenotypes on neural cell populations. Specific highlighted examples include engineered biomaterials and microfluidic platforms for spatiotemporal control over the presentation of morphogen gradients.
Collapse
Affiliation(s)
- Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
24
|
Lee UN, Day JH, Haack AJ, Bretherton RC, Lu W, DeForest CA, Theberge AB, Berthier E. Layer-by-layer fabrication of 3D hydrogel structures using open microfluidics. LAB ON A CHIP 2020; 20:525-536. [PMID: 31915779 PMCID: PMC8018606 DOI: 10.1039/c9lc00621d] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Patterned deposition and 3D fabrication techniques have enabled the use of hydrogels for a number of applications including microfluidics, sensors, separations, and tissue engineering in which form fits function. Devices such as reconfigurable microvalves or implantable tissues have been created using lithography or casting techniques. Here, we present a novel open-microfluidic patterning method that utilizes surface tension forces to form hydrogel layers on top of each other, into a patterned 3D structure. We use a patterning device to form a temporary open microfluidic channel on an existing gel layer, allowing the controlled flow of unpolymerized gel in device-regions. After layer gelation and device removal, the process can be repeated iteratively to create multi-layered 3D structures. The use of open-microfluidic and surface tension-based methods to define the shape of each individual layer enables patterning to be performed with a simple pipette and with minimal dead-volume. Our method is compatible with unmodified (native) biological hydrogels, and other non-biological materials with precursor fluid properties compatible with capillary flow. With our open-microfluidic layer-by-layer fabrication method, we demonstrate the capability to build agarose, type I collagen, and polymer-peptide 3D structures featuring asymmetric designs, multiple components, overhanging features, and cell-laden regions.
Collapse
Affiliation(s)
- Ulri N Lee
- Department of Chemistry, University of Washington, Seattle, WA, USA.
| | - John H Day
- Department of Chemistry, University of Washington, Seattle, WA, USA.
| | - Amanda J Haack
- Department of Chemistry, University of Washington, Seattle, WA, USA. and Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Ross C Bretherton
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| | - Wenbo Lu
- Department of Chemistry, University of Washington, Seattle, WA, USA.
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, WA, USA. and Department of Chemical Engineering, University of Washington, Seattle, WA, USA and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, USA
| | - Ashleigh B Theberge
- Department of Chemistry, University of Washington, Seattle, WA, USA. and Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|
25
|
Li Y, Motschman JD, Kelly ST, Yellen BB. Injection Molded Microfluidics for Establishing High-Density Single Cell Arrays in an Open Hydrogel Format. Anal Chem 2020; 92:2794-2801. [PMID: 31934750 PMCID: PMC7295173 DOI: 10.1021/acs.analchem.9b05099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Here, we develop an injection molded microfluidic approach for single cell analysis by making use of (1) rapidly curing injectable hydrogels, (2) a high density microfluidic weir trap array, and (3) reversibly bonded PDMS lids that are strong enough to withstand the injection molding process, but which can be peeled off after the hydrogel sets. This approach allows for single cell patterns to be created with densities exceeding 40 cells per mm2, is amenable to high speed imaging, and creates microfluidic devices that enable efficient nutrient and gas exchange and the delivery of specific biological and chemical reagents to individual cells. We show that it is possible to organize up to 10 000 single cells in a few minutes on the device, and we developed an image analysis program to automatically analyze the single-cell capture efficiency. The results show single cell trapping rates were better than 80%. We also demonstrate that the genomic DNA of the single cells trapped in the hydrogel can be amplified via localized, multiple displacement amplification in a massively parallel format, which offers a promising strategy for analyzing single cell genomes. Finally, we show the ability to perform selective staining of individual cells with a commercial bioprinter, providing proof of concept of its ability to deliver tailored reagents to specific cells in an array for future downstream analysis. This injection molded microfluidic approach leverages the benefits of both closed and open microfluidics, allows multiday single cell cultures, direct access to the trapped cells for genotypic end point studies.
Collapse
Affiliation(s)
- Ying Li
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan National Laboratory for Optoelectronics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jeffrey D. Motschman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Sean T. Kelly
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Benjamin B. Yellen
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
26
|
Mofazzal Jahromi MA, Abdoli A, Rahmanian M, Bardania H, Bayandori M, Moosavi Basri SM, Kalbasi A, Aref AR, Karimi M, Hamblin MR. Microfluidic Brain-on-a-Chip: Perspectives for Mimicking Neural System Disorders. Mol Neurobiol 2019; 56:8489-8512. [PMID: 31264092 PMCID: PMC6842047 DOI: 10.1007/s12035-019-01653-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/15/2019] [Indexed: 01/09/2023]
Abstract
Neurodegenerative diseases (NDDs) include more than 600 types of nervous system disorders in humans that impact tens of millions of people worldwide. Estimates by the World Health Organization (WHO) suggest NDDs will increase by nearly 50% by 2030. Hence, development of advanced models for research on NDDs is needed to explore new therapeutic strategies and explore the pathogenesis of these disorders. Different approaches have been deployed in order to investigate nervous system disorders, including two-and three-dimensional (2D and 3D) cell cultures and animal models. However, these models have limitations, such as lacking cellular tension, fluid shear stress, and compression analysis; thus, studying the biochemical effects of therapeutic molecules on the biophysiological interactions of cells, tissues, and organs is problematic. The microfluidic "organ-on-a-chip" is an inexpensive and rapid analytical technology to create an effective tool for manipulation, monitoring, and assessment of cells, and investigating drug discovery, which enables the culture of various cells in a small amount of fluid (10-9 to 10-18 L). Thus, these chips have the ability to overcome the mentioned restrictions of 2D and 3D cell cultures, as well as animal models. Stem cells (SCs), particularly neural stem cells (NSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) have the capability to give rise to various neural system cells. Hence, microfluidic organ-on-a-chip and SCs can be used as potential research tools to study the treatment of central nervous system (CNS) and peripheral nervous system (PNS) disorders. Accordingly, in the present review, we discuss the latest progress in microfluidic brain-on-a-chip as a powerful and advanced technology that can be used in basic studies to investigate normal and abnormal functions of the nervous system.
Collapse
Affiliation(s)
- Mirza Ali Mofazzal Jahromi
- Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Abdoli
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Parasitology and Mycology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mohammad Rahmanian
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Anesthesiology, Critical Care, and Pain Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mehrdad Bayandori
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amir Reza Aref
- Department of Cancer Biology, Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Department of Genetics, Harvard Medical School, Boston, MA, 02215, USA
| | - Mahdi Karimi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Dermatology, Harvard Medical School, Boston, MA, USA.
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.
| |
Collapse
|
27
|
O’Grady BJ, Balikov DA, Lippmann ES, Bellan LM. Spatiotemporal Control of Morphogen Delivery to Pattern Stem Cell Differentiation in Three-Dimensional Hydrogels. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2019; 51:e97. [PMID: 31756050 PMCID: PMC6876696 DOI: 10.1002/cpsc.97] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Morphogens are biological molecules that alter cellular identity and behavior across both space and time. During embryonic development, morphogen spatial localization can be confined to small volumes in a single tissue or permeate throughout an entire organism, and the temporal effects of morphogens can range from fractions of a second to several days. In most cases, morphogens are presented as a gradient to adjacent cells within tissues to pattern cell fate. As such, to appropriately model development and build representative multicellular architectures in vitro, it is vital to recapitulate these gradients during stem cell differentiation. However, the ability to control morphogen presentation within in vitro systems remains challenging. Here, we describe an innovative platform using channels patterned within thick, three-dimensional hydrogels that deliver multiple morphogens to embedded cells, thereby demonstrating exquisite control over both spatial and temporal variations in morphogen presentation. This generalizable approach should have broad utility for researchers interested in patterning in vitro tissue structures. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Brian J. O’Grady
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
- BJO and DAB contributed equally to this work as co-first authors
| | - Daniel A. Balikov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- BJO and DAB contributed equally to this work as co-first authors
| | - Ethan S. Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- ESL and LMB contributed equally to this work as co-senior authors
| | - Leon M. Bellan
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- ESL and LMB contributed equally to this work as co-senior authors
| |
Collapse
|
28
|
Kim JA, Hong S, Rhee WJ. Microfluidic three-dimensional cell culture of stem cells for high-throughput analysis. World J Stem Cells 2019; 11:803-816. [PMID: 31693013 PMCID: PMC6828593 DOI: 10.4252/wjsc.v11.i10.803] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/02/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
Although the recent advances in stem cell engineering have gained a great deal of attention due to their high potential in clinical research, the applicability of stem cells for preclinical screening in the drug discovery process is still challenging due to difficulties in controlling the stem cell microenvironment and the limited availability of high-throughput systems. Recently, researchers have been actively developing and evaluating three-dimensional (3D) cell culture-based platforms using microfluidic technologies, such as organ-on-a-chip and organoid-on-a-chip platforms, and they have achieved promising breakthroughs in stem cell engineering. In this review, we start with a comprehensive discussion on the importance of microfluidic 3D cell culture techniques in stem cell research and their technical strategies in the field of drug discovery. In a subsequent section, we discuss microfluidic 3D cell culture techniques for high-throughput analysis for use in stem cell research. In addition, some potential and practical applications of organ-on-a-chip or organoid-on-a-chip platforms using stem cells as drug screening and disease models are highlighted.
Collapse
Affiliation(s)
- Jeong Ah Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, South Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon 34113, South Korea
| | - Soohyun Hong
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, South Korea
- Program in Biomicro System Technology, Korea University, Seoul 02841, South Korea
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon 22012, South Korea
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, South Korea
| |
Collapse
|
29
|
Li N, Xie T, Sun Y. Towards organogenesis and morphogenesis in vitro: harnessing engineered microenvironment and autonomous behaviors of pluripotent stem cells. Integr Biol (Camb) 2019; 10:574-586. [PMID: 30225509 DOI: 10.1039/c8ib00116b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, researchers have been attempting to control pluripotent stem cell fate or generate self-organized tissues from stem cells. Advances in bioengineering enable generation of organotypic structures, which capture the cellular components, spatial cell organization and even some functions of tissues or organs in development. However, only a few engineering tools have been utilized to regulate the formation and organization of spatially complex tissues derived from stem cells. Here, we provide a review of recent progress in the culture of organotypic structures in vitro, focusing on how microengineering approaches including geometric confinement, extracellular matrix (ECM) property modulation, spatially controlled biochemical factors, and external forces, can be utilized to generate organotypic structures. Moreover, we will discuss potential technologies that can be applied to further control both soluble and insoluble factors spatiotemporally in vitro. In summary, advanced engineered approaches have a great promise in generating miniaturized tissues and organs in a reproducible fashion, facilitating the cellular and molecular understanding of embryogenesis and morphogenesis processes.
Collapse
Affiliation(s)
- Ningwei Li
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | | | | |
Collapse
|
30
|
Regier MC, Olszewski E, Carter CC, Aitchison JD, Kaushansky A, Davis J, Berthier E, Beebe DJ, Stevens KR. Spatial presentation of biological molecules to cells by localized diffusive transfer. LAB ON A CHIP 2019; 19:2114-2126. [PMID: 31111131 PMCID: PMC6755031 DOI: 10.1039/c9lc00122k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Cellular decisions in human development, homeostasis, regeneration, and disease are coordinated in large part by signals that are spatially localized in tissues. These signals are often soluble, such that biomolecules produced by one cell diffuse to receiving cells. To recapitulate soluble factor patterning in vitro, several microscale strategies have been developed. However, these techniques often introduce new variables into cell culture experiments (e.g., fluid flow) or are limited in their ability to pattern diverse solutes in a user-defined manner. To address these challenges, we developed an adaptable method that facilitates spatial presentation of biomolecules across cells in traditional open cultures in vitro. This technique employs device inserts that are placed in standard culture wells, which support localized diffusive pattern transmission through microscale spaces between device features and adherent cells. Devices can be removed and cultures can be returned to standard media following patterning. We use this method to spatially control cell labeling with pattern features ranging in scale from several hundred microns to millimeters and with sequential application of multiple patterns. To better understand the method we investigate relationships between pattern fidelity, device geometry, and consumption and diffusion kinetics using finite element modeling. We then apply the method to spatially defining reporter cell heterogeneity by patterning a small molecule modulator of genetic recombination with the requisite sustained exposure. Finally, we demonstrate use of this method for patterning larger and more slowly diffusing particles by creating focal sites of gene delivery and infection with adenoviral, lentiviral, and Zika virus particles. Thus, our method leverages devices that interface with standard culture vessels to pattern diverse diffusible factors, geometries, exposure dynamics, and recipient cell types, making it well poised for adoption by researchers across various fields of biological research.
Collapse
Affiliation(s)
- Mary C Regier
- Department of Bioengineering, University of Washington, 98195 Seattle, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Regier MC, Tokar JJ, Warrick JW, Pabon L, Berthier E, Beebe DJ, Stevens KR. User-defined morphogen patterning for directing human cell fate stratification. Sci Rep 2019; 9:6433. [PMID: 31015521 PMCID: PMC6478938 DOI: 10.1038/s41598-019-42874-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Concentration gradients of biochemical stimuli such as morphogens play a critical role in directing cell fate patterning across species and throughout development but are not commonly recapitulated in vitro. While in vitro biomolecule gradients have been generated using customized microfluidic platforms, broad implementation has been limited because these platforms introduce new variables to cell culture such as externally driven flow, culture in a specialized matrix, or extended time for in situ long range diffusion. Here we introduce a method that enables preforming and then transferring user-controlled gradients to cells in standard "open" cultures. Our gradient patterning devices are modular and decoupled from the culture substrate. We find that gradient generation and transfer are predictable by finite element modeling and that device and loading parameters can be used to tune the stimulus pattern. Furthermore, we demonstrate use of these devices to spatially define morphogen signal gradients and direct peri-gastrulation fate stratification of human pluripotent stem cells. This method for extrinsic application of biochemical signal gradients can thus be used to spatially influence cellular fate decisions in a user-controlled manner.
Collapse
Affiliation(s)
- Mary C Regier
- Department of Bioengineering, University of Washington, 98195, Seattle, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jacob J Tokar
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin - Madison, 53705, Madison, USA
| | - Lil Pabon
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Pathology, University of Washington, 98195, Seattle, USA
| | - Erwin Berthier
- Department of Chemistry, University of Washington, 98195, Seattle, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, 98195, Seattle, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA.
- Department of Pathology, University of Washington, 98195, Seattle, USA.
| |
Collapse
|
32
|
Samal P, van Blitterswijk C, Truckenmüller R, Giselbrecht S. Grow with the Flow: When Morphogenesis Meets Microfluidics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805764. [PMID: 30767289 DOI: 10.1002/adma.201805764] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Developmental biology has advanced the understanding of the intricate and dynamic processes involved in the formation of an organism from a single cell. However, many gaps remain in the knowledge of embryonic development, especially regarding tissue morphogenesis. A possible approach to mimic such phenomena uses pluripotent stem cells in in vitro morphogenetic models. Herein, these systems are summarized with emphasis on the ability to better manipulate and control cellular interfaces with either liquid or solid materials using microengineered tools, which is critical for attaining deeper insights into pattern formation and stem cell differentiation during organogenesis. The role of conventional and customized cell-culture systems in supporting important advances in the field of morphogenesis is discussed, and the fascinating role that material sciences and microengineering currently play and are expected to play in the future is highlighted. In conclusion, it is proffered that continued microfluidics innovations when applied to morphogenesis promise to provide important insights to advance many multidisciplinary fields, including regenerative medicine.
Collapse
Affiliation(s)
- Pinak Samal
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Clemens van Blitterswijk
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Roman Truckenmüller
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
33
|
Cui P, Wang S. Application of microfluidic chip technology in pharmaceutical analysis: A review. J Pharm Anal 2018; 9:238-247. [PMID: 31452961 PMCID: PMC6704040 DOI: 10.1016/j.jpha.2018.12.001] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 01/18/2023] Open
Abstract
The development of pharmaceutical analytical methods represents one of the most significant aspects of drug development. Recent advances in microfabrication and microfluidics could provide new approaches for drug analysis, including drug screening, active testing and the study of metabolism. Microfluidic chip technologies, such as lab-on-a-chip technology, three-dimensional (3D) cell culture, organs-on-chip and droplet techniques, have all been developed rapidly. Microfluidic chips coupled with various kinds of detection techniques are suitable for the high-throughput screening, detection and mechanistic study of drugs. This review highlights the latest (2010–2018) microfluidic technology for drug analysis and discusses the potential future development in this field.
Collapse
Affiliation(s)
- Ping Cui
- School of Pharmacy, Xi'an Jiaotong University Health Science Center, #76, Yanta West Road, Xi'an 710061, China.,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an 710061, China
| | - Sicen Wang
- School of Pharmacy, Xi'an Jiaotong University Health Science Center, #76, Yanta West Road, Xi'an 710061, China.,Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an 710061, China
| |
Collapse
|
34
|
Akay M, Hite J, Avci NG, Fan Y, Akay Y, Lu G, Zhu JJ. Drug Screening of Human GBM Spheroids in Brain Cancer Chip. Sci Rep 2018; 8:15423. [PMID: 30337660 PMCID: PMC6194126 DOI: 10.1038/s41598-018-33641-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), an extremely invasive and high-grade (grade IV) glioma, is the most common and aggressive form of brain cancer. It has a poor prognosis, with a median overall survival of only 11 months in the general GBM population and 14.6 to 21 months in clinical trial participants with standard GBM therapies, including maximum safe craniotomy, adjuvant radiation, and chemotherapies. Therefore, new approaches for developing effective treatments, such as a tool for assessing tumor cell drug response before drug treatments are administered, are urgently needed to improve patient survival. To address this issue, we developed an improved brain cancer chip with a diffusion prevention mechanism that blocks drugs crossing from one channel to another. In the current study, we demonstrate that the chip has the ability to culture 3D spheroids from patient tumor specimen-derived GBM cells obtained from three GBM patients. Two clinical drugs used to treat GBM, temozolomide (TMZ) and bevacizumab (Avastin, BEV), were applied and a range of relative concentrations was generated by the microfluidic channels in the brain cancer chip. The results showed that TMZ works more effectively when used in combination with BEV compared to TMZ alone. We believe that this low-cost brain cancer chip could be further developed to generate optimal combination of chemotherapy drugs tailored to individual GBM patients.
Collapse
Affiliation(s)
- Metin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA.
| | - John Hite
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Naze Gul Avci
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yantao Fan
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yasemin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Guangrong Lu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| | - Jay-Jiguang Zhu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| |
Collapse
|
35
|
Towards Three-Dimensional Dynamic Regulation and In Situ Characterization of Single Stem Cell Phenotype Using Microfluidics. Mol Biotechnol 2018; 60:843-861. [DOI: 10.1007/s12033-018-0113-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Wasserberg D, Zhang X, Breukers C, Connell BJ, Baeten E, van den Blink D, S O L À Benet È, Bloem AC, Nijhuis M, Wensing AMJ, Terstappen LWMM, Beck M. All-printed cell counting chambers with on-chip sample preparation for point-of-care CD4 counting. Biosens Bioelectron 2018; 117:659-668. [PMID: 30005387 DOI: 10.1016/j.bios.2018.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/27/2018] [Accepted: 07/01/2018] [Indexed: 12/19/2022]
Abstract
We demonstrate the fabrication of fully printed microfluidic CD4 counting chips with complete on-chip sample preparation and their applicability as a CD4 counting assay using samples from healthy donors and HIV-infected patients. CD4 counting in low-income and resource-limited point-of-care settings is only practical and affordable, if disposable tests can be fabricated at very low cost and all manual sample preparation is avoided, while operation as well as quantification is fully automated and independent of the skills of the operator. Here, we show the successful use of (inkjet) printing methods both to fabricate microfluidic cell counting chambers with controlled heights, and to deposit hydrogel layers with embedded fluorophore-labeled antibodies for on-chip sample preparation and reagent storage. The maturation process of gelatin after deposition prevents antibody wash-off during blood inflow very well, while temperature-controlled dissolution of the matrix ensures complete antibody release for immunostaining after the inflow has stopped. The prevention of antibody wash-off together with the subsequent complete antibody release guarantees a homogeneous fluorescence background, making rapid and accurate CD4 counting possible. We show the successful application of our fully printed CD4 counting chips on samples from healthy donors as well as from HIV-infected patients and find an excellent agreement between results from our method and from the gold standard, flow cytometry, in both cases.
Collapse
Affiliation(s)
- Dorothee Wasserberg
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Xichen Zhang
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Christian Breukers
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Bridgette J Connell
- University Medical Center Utrecht, Department of Medical Microbiology, Virology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Ellen Baeten
- University Medical Center Utrecht, Laboratory of Translational Immunology, Section Diagnostics, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Dorine van den Blink
- University Medical Center Utrecht, Laboratory of Translational Immunology, Section Diagnostics, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Èlia S O L À Benet
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Andries C Bloem
- University Medical Center Utrecht, Laboratory of Translational Immunology, Section Diagnostics, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Monique Nijhuis
- University Medical Center Utrecht, Department of Medical Microbiology, Virology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Annemarie M J Wensing
- University Medical Center Utrecht, Department of Medical Microbiology, Virology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Leon W M M Terstappen
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Markus Beck
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands.
| |
Collapse
|
37
|
Kim YT, Castro K, Bhattacharjee N, Folch A. Digital Manufacturing of Selective Porous Barriers in Microchannels Using Multi-Material Stereolithography. MICROMACHINES 2018; 9:E125. [PMID: 30424059 PMCID: PMC6187461 DOI: 10.3390/mi9030125] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 01/09/2023]
Abstract
We have developed a sequential stereolithographic co-printing process using two different resins for fabricating porous barriers in microfluidic devices. We 3D-printed microfluidic channels with a resin made of poly(ethylene glycol) diacrylate (MW = 258) (PEG-DA-258), a UV photoinitiator, and a UV sensitizer. The porous barriers were created within the microchannels in a different resin made of either PEG-DA (MW = 575) (PEG-DA-575) or 40% (w/w in water) PEG-DA (MW = 700) (40% PEG-DA-700). We showed selective hydrogen ion diffusion across a 3D-printed PEG-DA-575 porous barrier in a cross-channel diffusion chip by observing color changes in phenol red, a pH indicator. We also demonstrated the diffusion of fluorescein across a 3D-printed 40% PEG-DA-700 porous barrier in a symmetric-channel diffusion chip by measuring fluorescence intensity changes across the porous barrier. Creating microfluidic chips with integrated porous barriers using a semi-automated 3D printing process shortens the design and processing time, avoids assembly and bonding complications, and reduces manufacturing costs compared to micromolding processes. We believe that our digital manufacturing method for fabricating selective porous barriers provides an inexpensive, simple, convenient and reproducible route to molecule delivery in the fields of molecular filtration and cell-based microdevices.
Collapse
|
38
|
Martins JP, Torrieri G, Santos HA. The importance of microfluidics for the preparation of nanoparticles as advanced drug delivery systems. Expert Opin Drug Deliv 2018; 15:469-479. [PMID: 29508630 DOI: 10.1080/17425247.2018.1446936] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Nanoparticles are anticipated to overcome persistent challenges in efficient drug delivery, but the limitations associated with conventional methods of preparation are resulting in slow translation from research to clinical applications. Due to their enormous potential, microfluidic technologies have emerged as an advanced approach for the development of drug delivery systems with well-defined physicochemical characteristics and in a reproducible manner. AREAS COVERED This review provides an overview of microfluidic devices and materials used for their manufacturing, together with the flow patterns and regimes commonly used for nanoparticle preparation. Additionally, the different geometries used in droplet microfluidics are reviewed, with particular attention to the co-flow geometry used for the production of nanoparticles. Finally, this review summarizes the main and most recent nanoparticulate systems prepared using microfluidics, including drug nanosuspensions, polymeric, lipid, structured, and theranostic nanoparticles. EXPERT OPINION The production of nanoparticles at industrial scale is still a challenge, but the microfluidic technologies bring exciting opportunities to develop drug delivery systems that can be engineered in an easy, cost-effective and reproducible manner. As a highly interdisciplinary research field, more efforts and general acceptance are needed to allow for the translation of nanoparticulate drug delivery systems from academic research to the clinical practice.
Collapse
Affiliation(s)
- João Pedro Martins
- a Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy , University of Helsinki , Helsinki , Finland
| | - Giulia Torrieri
- a Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy , University of Helsinki , Helsinki , Finland
| | - Hélder A Santos
- a Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy , University of Helsinki , Helsinki , Finland.,b Helsinki Institute of Life Science (HiLIFE) , University of Helsinki , Helsinki , Finland
| |
Collapse
|
39
|
Osaki T, Shin Y, Sivathanu V, Campisi M, Kamm RD. In Vitro Microfluidic Models for Neurodegenerative Disorders. Adv Healthc Mater 2018; 7. [PMID: 28881425 DOI: 10.1002/adhm.201700489] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/18/2017] [Indexed: 01/09/2023]
Abstract
Microfluidic devices enable novel means of emulating neurodegenerative disease pathophysiology in vitro. These organ-on-a-chip systems can potentially reduce animal testing and substitute (or augment) simple 2D culture systems. Reconstituting critical features of neurodegenerative diseases in a biomimetic system using microfluidics can thereby accelerate drug discovery and improve our understanding of the mechanisms of several currently incurable diseases. This review describes latest advances in modeling neurodegenerative diseases in the central nervous system and the peripheral nervous system. First, this study summarizes fundamental advantages of microfluidic devices in the creation of compartmentalized cell culture microenvironments for the co-culture of neurons, glial cells, endothelial cells, and skeletal muscle cells and in their recapitulation of spatiotemporal chemical gradients and mechanical microenvironments. Then, this reviews neurodegenerative-disease-on-a-chip models focusing on Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Finally, this study discusses about current drawbacks of these models and strategies that may overcome them. These organ-on-chip technologies can be useful to be the first line of testing line in drug development and toxicology studies, which can contribute significantly to minimize the phase of animal testing steps.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Vivek Sivathanu
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Marco Campisi
- Department of Mechanical and Aerospace EngineeringPolitecnico di Torino Corso Duca degli Abruzzi 24 10129 Torino Italy
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
- Department of Biological EngineeringMassachusetts Institutes of Technology 500 Technology Square, MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| |
Collapse
|
40
|
Geraili A, Jafari P, Hassani MS, Araghi BH, Mohammadi MH, Ghafari AM, Tamrin SH, Modarres HP, Kolahchi AR, Ahadian S, Sanati-Nezhad A. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms. Adv Healthc Mater 2018; 7. [PMID: 28910516 DOI: 10.1002/adhm.201700426] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/01/2017] [Indexed: 01/09/2023]
Abstract
Organ-on-chip (OOC) platforms have attracted attentions of pharmaceutical companies as powerful tools for screening of existing drugs and development of new drug candidates. OOCs have primarily used human cell lines or primary cells to develop biomimetic tissue models. However, the ability of human stem cells in unlimited self-renewal and differentiation into multiple lineages has made them attractive for OOCs. The microfluidic technology has enabled precise control of stem cell differentiation using soluble factors, biophysical cues, and electromagnetic signals. This study discusses different tissue- and organ-on-chip platforms (i.e., skin, brain, blood-brain barrier, bone marrow, heart, liver, lung, tumor, and vascular), with an emphasis on the critical role of stem cells in the synthesis of complex tissues. This study further recaps the design, fabrication, high-throughput performance, and improved functionality of stem-cell-based OOCs, technical challenges, obstacles against implementing their potential applications, and future perspectives related to different experimental platforms.
Collapse
Affiliation(s)
- Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
| | - Parya Jafari
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
- Department of Electrical Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohsen Sheikh Hassani
- Department of Systems and Computer Engineering; Carleton University; 1125 Colonel By Drive Ottawa K1S 5B6 ON Canada
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Mohammad Ghafari
- Department of Stem Cells and Developmental Biology; Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology; Tehran 16635-148 Iran
| | - Sara Hasanpour Tamrin
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
- Center for Bioengineering Research and Education; Biomedical Engineering Program; University of Calgary; Calgary T2N 1N4 AB Canada
| |
Collapse
|
41
|
Compartmentalized Microfluidic Platforms: The Unrivaled Breakthrough of In Vitro Tools for Neurobiological Research. J Neurosci 2017; 36:11573-11584. [PMID: 27852766 DOI: 10.1523/jneurosci.1748-16.2016] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/08/2016] [Accepted: 09/28/2016] [Indexed: 12/15/2022] Open
Abstract
Microfluidic technology has become a valuable tool to the scientific community, allowing researchers to study fine cellular mechanisms with higher variable control compared with conventional systems. It has evolved tremendously, and its applicability and flexibility made its usage grow exponentially and transversely to several research fields. This has been particularly noticeable in neuroscience research, where microfluidic platforms made it possible to address specific questions extending from axonal guidance, synapse formation, or axonal transport to the development of 3D models of the CNS to allow pharmacological testing and drug screening. Furthermore, the continuous upgrade of microfluidic platforms has allowed a deeper study of the communication occurring between different neuronal and glial cells or between neurons and other peripheral tissues, both in physiological and pathological conditions. Importantly, the evolution of microfluidic technology has always been accompanied by the development of new computational tools addressing data acquisition, analysis, and modeling.
Collapse
|
42
|
Liu Y, Lu H. Microfluidics in systems biology-hype or truly useful? Curr Opin Biotechnol 2017; 39:215-220. [PMID: 27267565 DOI: 10.1016/j.copbio.2016.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/13/2022]
Abstract
Systems biology often relies on large-scale measurements and model-building to understand how complex biological systems function. Microfluidic technology has been touted as a tool for high-throughput experiments and has been a valuable tool to some systems biology research. This review focuses on applications where microfluidics can enhance experimental sensitivity and throughput, particularly in recent development in single-cell analyses and analyses on multi-cellular or complex biological entities. We conclude that microfluidics is not necessarily always useful for systems biology, but when used appropriately can greatly enhance experimentalists' ability to measure and control, and thereby enhance the understanding of and expand the utility of biological systems.
Collapse
Affiliation(s)
- Yi Liu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, United States
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, United States.
| |
Collapse
|
43
|
Ezra Tsur E, Zimerman M, Maor I, Elrich A, Nahmias Y. Microfluidic Concentric Gradient Generator Design for High-Throughput Cell-Based Studies. Front Bioeng Biotechnol 2017; 5:21. [PMID: 28447031 PMCID: PMC5388772 DOI: 10.3389/fbioe.2017.00021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/17/2017] [Indexed: 01/22/2023] Open
Abstract
Gradients of diffusible signaling molecules play important role in various processes, ranging from cell differentiation to toxicological evaluation. Microfluidic technology provides an accurate control of tempospatial conditions. However, current microfluidic platforms are not designed to handle multiple gradients and cell populations simultaneously. Here, we demonstrate a rapidly adaptable microfluidic design able to expose multiple cell populations to an array of chemical gradients. Our design is based on pressure-equilibrated concentric channels and a pressure-dissipating control layer, facilitating the seeding of multiple cell populations in a single device. The design was numerically evaluated and experimentally validated. The device consists of 8 radiating stimuli channels and 12 circular cell culture channels, creating an array of 96 different continuous gradients that can be simultaneously monitored over time.
Collapse
Affiliation(s)
- Elishai Ezra Tsur
- Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Neuro-Biomorphic Engineering Lab, Faculty of Engineering, Jerusalem College of Technology, Jerusalem, Israel
| | - Michal Zimerman
- Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Idan Maor
- Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avner Elrich
- Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yaakov Nahmias
- Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
44
|
Multiscale microenvironmental perturbation of pluripotent stem cell fate and self-organization. Sci Rep 2017; 7:44711. [PMID: 28303935 PMCID: PMC5356187 DOI: 10.1038/srep44711] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/13/2017] [Indexed: 01/09/2023] Open
Abstract
The combination of microfluidics with engineered three-dimensional (3D) matrices can bring new insights into the fate regulation of stem cells and their self-organization into organoids. Although there has been progress in 3D stem cell culturing, most existing in vitro methodologies do not allow for mimicking of the spatiotemporal heterogeneity of stimuli that drive morphogenetic processes in vivo. To address this, we present a perfusion-free microchip concept for the in vitro 3D perturbation of stem cell fate. Stem cells are encapsulated in a hydrogel compartment that is flanked by open reservoirs for the diffusion-driven generation of biomolecule gradients. Juxtaposing additional compartments bearing supportive cells enables investigating the influence of long range cell-cell communication. We explore the utility of the microchips in manipulating early fate choices and self-organizing characteristics of 3D-cultured mouse embryonic stem cells (mESCs) under neural differentiation conditions and exposure to gradients of leukemia inhibitory factor (LIF). mESCs respond to LIF gradients in a spatially dependent manner. At higher LIF concentrations, multicellular colonies maintain pluripotency in contrast, at lower concentrations, mESCs develop into apicobasally polarized epithelial cysts. This versatile system can help to systematically explore the role of multifactorial microenvironments in promoting self-patterning of various stem cell types.
Collapse
|
45
|
Jungnickel C, Tsurkan MV, Wogan K, Werner C, Schlierf M. Bottom-Up Structuring and Site-Selective Modification of Hydrogels Using a Two-Photon [2+2] Cycloaddition of Maleimide. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1603327. [PMID: 27862380 DOI: 10.1002/adma.201603327] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/26/2016] [Indexed: 06/06/2023]
Abstract
Creating hydrogel systems to mimic the extracellular matrix is often limited by their static nature. The use of a two-photon [2+2] cycloaddition of maleimide groups to structure surfaces, to create hydrogels, and add 3D modifications with sub-micrometer precision is reported. The absence of photoinitiators and usage of near-infrared light is promising for future in vivo studies.
Collapse
Affiliation(s)
- Christiane Jungnickel
- B CUBE - Center for Molecular Bioengineering, Technische Universität Dresden, Arnoldstraße 18, 01307, Dresden, Germany
- Leibniz-Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Straße 6, 01069, Dresden, Germany
| | - Mikhail V Tsurkan
- Leibniz-Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Straße 6, 01069, Dresden, Germany
| | - Kristin Wogan
- B CUBE - Center for Molecular Bioengineering, Technische Universität Dresden, Arnoldstraße 18, 01307, Dresden, Germany
| | - Carsten Werner
- Leibniz-Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Straße 6, 01069, Dresden, Germany
| | - Michael Schlierf
- B CUBE - Center for Molecular Bioengineering, Technische Universität Dresden, Arnoldstraße 18, 01307, Dresden, Germany
| |
Collapse
|
46
|
Krishna L, Dhamodaran K, Jayadev C, Chatterjee K, Shetty R, Khora SS, Das D. Nanostructured scaffold as a determinant of stem cell fate. Stem Cell Res Ther 2016; 7:188. [PMID: 28038681 PMCID: PMC5203716 DOI: 10.1186/s13287-016-0440-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The functionality of stem cells is tightly regulated by cues from the niche, comprising both intrinsic and extrinsic cell signals. Besides chemical and growth factors, biophysical signals are important components of extrinsic signals that dictate the stem cell properties. The materials used in the fabrication of scaffolds provide the chemical cues whereas the shape of the scaffolds provides the biophysical cues. The effect of the chemical composition of the scaffolds on stem cell fate is well researched. Biophysical signals such as nanotopography, mechanical forces, stiffness of the matrix, and roughness of the biomaterial influence the fate of stem cells. However, not much is known about their role in signaling crosstalk, stem cell maintenance, and directed differentiation. Among the various techniques for scaffold design, nanotechnology has special significance. The role of nanoscale topography in scaffold design for the regulation of stem cell behavior has gained importance in regenerative medicine. Nanotechnology allows manipulation of highly advanced surfaces/scaffolds for optimal regulation of cellular behavior. Techniques such as electrospinning, soft lithography, microfluidics, carbon nanotubes, and nanostructured hydrogel are described in this review, along with their potential usage in regenerative medicine. We have also provided a brief insight into the potential signaling crosstalk that is triggered by nanomaterials that dictate a specific outcome of stem cells. This concise review compiles recent developments in nanoscale architecture and its importance in directing stem cell differentiation for prospective therapeutic applications.
Collapse
Affiliation(s)
- Lekshmi Krishna
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.,School of Bioscience and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Kamesh Dhamodaran
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.,School of Bioscience and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Chaitra Jayadev
- Vitreoretina Services, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore, Karnataka, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - S S Khora
- School of Bioscience and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Debashish Das
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.
| |
Collapse
|
47
|
Freudenberg U, Liang Y, Kiick KL, Werner C. Glycosaminoglycan-Based Biohybrid Hydrogels: A Sweet and Smart Choice for Multifunctional Biomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:8861-8891. [PMID: 27461855 PMCID: PMC5152626 DOI: 10.1002/adma.201601908] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 05/30/2016] [Indexed: 05/12/2023]
Abstract
Glycosaminoglycans (GAGs) govern important functional characteristics of the extracellular matrix (ECM) in living tissues. Incorporation of GAGs into biomaterials opens up new routes for the presentation of signaling molecules, providing control over development, homeostasis, inflammation, and tumor formation and progression. Recent approaches to GAG-based materials are reviewed, highlighting the formation of modular, tunable biohybrid hydrogels by covalent and non-covalent conjugation schemes, including both theory-driven design concepts and advanced processing technologies. Examples of the application of the resulting materials in biomedical studies are provided. For perspective, solid-phase and chemoenzymatic oligosaccharide synthesis methods for GAG-derived motifs, rational and high-throughput design strategies for GAG-based materials, and the utilization of the factor-scavenging characteristics of GAGs are highlighted.
Collapse
Affiliation(s)
- Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Hohe Str. 6, 01069 Dresden, Germany
| | - Yingkai Liang
- Department of Materials Science and Engineering and Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States,
| | - Kristi L. Kiick
- Department of Materials Science and Engineering and Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States and Delaware Biotechnology Institute, 15 Innovation Way, Newark, Delaware 19716, United States
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden (IPF), Max Bergmann Center of Biomaterials Dresden (MBC), Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Hohe Str. 6, 01069 Dresden, Germany
| |
Collapse
|
48
|
Peng H, Rübsam K, Jakob F, Schwaneberg U, Pich A. Tunable Enzymatic Activity and Enhanced Stability of Cellulase Immobilized in Biohybrid Nanogels. Biomacromolecules 2016; 17:3619-3631. [DOI: 10.1021/acs.biomac.6b01119] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Huan Peng
- DWI-Leibniz Institute
for Interactive Materials e.V., Aachen, Germany
| | - Kristin Rübsam
- DWI-Leibniz Institute
for Interactive Materials e.V., Aachen, Germany
| | - Felix Jakob
- DWI-Leibniz Institute
for Interactive Materials e.V., Aachen, Germany
| | | | - Andrij Pich
- DWI-Leibniz Institute
for Interactive Materials e.V., Aachen, Germany
| |
Collapse
|
49
|
Eltaher HM, Yang J, Shakesheff KM, Dixon JE. Highly efficient intracellular transduction in three-dimensional gradients for programming cell fate. Acta Biomater 2016; 41:181-92. [PMID: 27265151 DOI: 10.1016/j.actbio.2016.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/22/2016] [Accepted: 06/02/2016] [Indexed: 01/01/2023]
Abstract
UNLABELLED Fundamental behaviour such as cell fate, growth and death are mediated through the control of key genetic transcriptional regulators. These regulators are activated or repressed by the integration of multiple signalling molecules in spatio-temporal gradients. Engineering these gradients is complex but considered key in controlling tissue formation in regenerative medicine approaches. Direct programming of cells using exogenously delivered transcription factors can by-pass growth factor complexity but there is still a requirement to deliver such activity spatio-temporally. We previously developed a technology termed GAG-binding enhanced transduction (GET) to efficiently deliver a variety of cargoes intracellularly using GAG-binding domains to promote cell targeting, and cell penetrating peptides (CPPs) to allow cell entry. Herein we demonstrate that GET can be used in a three dimensional (3D) hydrogel matrix to produce gradients of intracellular transduction of mammalian cells. Using a compartmentalised diffusion model with a source-gel-sink (So-G-Si) assembly, we created gradients of reporter proteins (mRFP1-tagged) and a transcription factor (TF, myogenic master regulator MyoD) and showed that GET can be used to deliver molecules into cells spatio-temporally by monitoring intracellular transduction and gene expression programming as a function of location and time. The ability to spatio-temporally control the intracellular delivery of functional proteins will allow the establishment of gradients of cell programming in hydrogels and approaches to direct cellular behaviour for many regenerative medicine applications. STATEMENT OF SIGNIFICANCE Regenerative medicine aims to reform functional biological tissues by controlling cell behaviour. Growth factors (GFs) are soluble cues presented to cells in spatio-temporal gradients and play important roles programming cell fate and gene expression. The efficient transduction of cells by GET (Glycosaminoglycan-enhanced transducing)-tagged transcription factors (TFs) can be used to by-pass GF-stimulation and directly program cells. For the first time we demonstrate diffusion of GET proteins generate stable protein transduction gradients. We demonstrated the feasibility of creating spatio-temporal gradients of GET-MyoD and show differential programing of myogenic differentiation. We believe that GET could provide a powerful tool to program cell behaviour using gradients of recombinant proteins that allow tissue generation directly by programming gene expression with TFs.
Collapse
|
50
|
González-Díaz EC, Varghese S. Hydrogels as Extracellular Matrix Analogs. Gels 2016; 2:E20. [PMID: 30674152 PMCID: PMC6318624 DOI: 10.3390/gels2030020] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/29/2016] [Accepted: 07/25/2016] [Indexed: 02/02/2023] Open
Abstract
The extracellular matrix (ECM) is the non-cellular component of tissue that provides physical scaffolding to cells. Emerging studies have shown that beyond structural support, the ECM provides tissue-specific biochemical and biophysical cues that are required for tissue morphogenesis and homeostasis. Hydrogel-based platforms have played a key role in advancing our knowledge of the role of ECM in regulating various cellular functions. Synthetic hydrogels allow for tunable biofunctionality, as their material properties can be tailored to mimic those of native tissues. This review discusses current advances in the design of hydrogels with defined physical and chemical properties. We also highlight research findings that demonstrate the impact of matrix properties on directing stem cell fate, such as self-renewal and differentiation. Recent and future efforts towards understanding cell-material interactions will not only advance our basic understanding, but will also help design tissue-specific matrices and delivery systems to transplant stem cells and control their response in vivo.
Collapse
Affiliation(s)
- Eva C González-Díaz
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Shyni Varghese
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|