1
|
Roehm KD, Chiesa I, Haithcock D, Gottardi R, Prabhakarpandian B. A vascularized microfluidic model of the osteochondral unit for modeling inflammatory response and therapeutic screening. LAB ON A CHIP 2024. [PMID: 39715348 DOI: 10.1039/d4lc00651h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Osteoarthritis (OA) has long been considered a disease of the articular cartilage. Within the past decade it has become increasingly clear that OA is a disease of the entire joint space and that interactions between articular cartilage and subchondral bone likely play an important role in the disease. Driven by this knowledge, we have created a novel microphysiological model of the osteochondral unit containing synovium, cartilage, bone, and vasculature in separate compartments with molecular and direct cell-cell interaction between the cells from the different tissue types. We have characterized the model in terms of differentiation by molecule and matrix secretion and shown that it demonstrates morphology and functionality that mimic the native characteristic of the joint space. Finally, we induced inflammation and subsequently rescued the model constructs by a known compound as proof of concept for anti-inflammatory drug screening applications.
Collapse
Affiliation(s)
- Kevin D Roehm
- CFD Research Corporation, 6820 Moquin Dr. N.W., Huntsville, AL 35806, USA.
| | - Irene Chiesa
- Department of Information Engineering and Research Center "Enrico Piaggio", University of Pisa, Italy
- Division of Otolaryngology, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dustin Haithcock
- CFD Research Corporation, 6820 Moquin Dr. N.W., Huntsville, AL 35806, USA.
| | - Riccardo Gottardi
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Division of Otolaryngology, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Pulmonary Medicine, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Ri.MED Foundation, Palermo, Italy
| | | |
Collapse
|
2
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [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: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
3
|
Babaliari E, Ranella A, Stratakis E. Microfluidic Systems for Neural Cell Studies. Bioengineering (Basel) 2023; 10:902. [PMID: 37627787 PMCID: PMC10451731 DOI: 10.3390/bioengineering10080902] [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: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Whereas the axons of the peripheral nervous system (PNS) spontaneously regenerate after an injury, the occurring regeneration is rarely successful because axons are usually directed by inappropriate cues. Therefore, finding successful ways to guide neurite outgrowth, in vitro, is essential for neurogenesis. Microfluidic systems reflect more appropriately the in vivo environment of cells in tissues such as the normal fluid flow within the body, consistent nutrient delivery, effective waste removal, and mechanical stimulation due to fluid shear forces. At the same time, it has been well reported that topography affects neuronal outgrowth, orientation, and differentiation. In this review, we demonstrate how topography and microfluidic flow affect neuronal behavior, either separately or in synergy, and highlight the efficacy of microfluidic systems in promoting neuronal outgrowth.
Collapse
Affiliation(s)
- Eleftheria Babaliari
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Anthi Ranella
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Emmanuel Stratakis
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
- Department of Physics, University of Crete, 70013 Heraklion, Greece
| |
Collapse
|
4
|
Yu L, Cavelier S, Hannon B, Wei M. Recent development in multizonal scaffolds for osteochondral regeneration. Bioact Mater 2023; 25:122-159. [PMID: 36817819 PMCID: PMC9931622 DOI: 10.1016/j.bioactmat.2023.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/30/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Osteochondral (OC) repair is an extremely challenging topic due to the complex biphasic structure and poor intrinsic regenerative capability of natural osteochondral tissue. In contrast to the current surgical approaches which yield only short-term relief of symptoms, tissue engineering strategy has been shown more promising outcomes in treating OC defects since its emergence in the 1990s. In particular, the use of multizonal scaffolds (MZSs) that mimic the gradient transitions, from cartilage surface to the subchondral bone with either continuous or discontinuous compositions, structures, and properties of natural OC tissue, has been gaining momentum in recent years. Scrutinizing the latest developments in the field, this review offers a comprehensive summary of recent advances, current hurdles, and future perspectives of OC repair, particularly the use of MZSs including bilayered, trilayered, multilayered, and gradient scaffolds, by bringing together onerous demands of architecture designs, material selections, manufacturing techniques as well as the choices of growth factors and cells, each of which possesses its unique challenges and opportunities.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Sacha Cavelier
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Brett Hannon
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Mei Wei
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Mechanical Engineering, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
5
|
Petta D, D'Amora U, D'Arrigo D, Tomasini M, Candrian C, Ambrosio L, Moretti M. Musculoskeletal tissues-on-a-chip: role of natural polymers in reproducing tissue-specific microenvironments. Biofabrication 2022; 14. [PMID: 35931043 DOI: 10.1088/1758-5090/ac8767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/05/2022] [Indexed: 11/12/2022]
Abstract
Over the past years, 3D in vitro models have been widely employed in the regenerative medicine field. Among them, organ-on-a-chip technology has the potential to elucidate cellular mechanism exploiting multichannel microfluidic devices to establish 3D co-culture systems that offer control over the cellular, physico-chemical and biochemical microenvironments. To deliver the most relevant cues to cells, it is of paramount importance to select the most appropriate matrix for mimicking the extracellular matrix of the native tissue. Natural polymers-based hydrogels are the elected candidates for reproducing tissue-specific microenvironments in musculoskeletal tissue-on-a-chip models owning to their interesting and peculiar physico-chemical, mechanical and biological properties. Despite these advantages, there is still a gap between the biomaterials complexity in conventional tissue engineering and the application of these biomaterials in 3D in vitro microfluidic models. In this review, the aim is to suggest the adoption of more suitable biomaterials, alternative crosslinking strategies and tissue engineered-inspired approaches in organ-on-a-chip to better mimic the complexity of physiological musculoskeletal tissues. Accordingly, after giving an overview of the musculoskeletal tissue compositions, the properties of the main natural polymers employed in microfluidic systems are investigated, together with the main musculoskeletal tissues-on-a-chip devices.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologis Lab, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Ugo D'Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, V.le J.F. Kennedy 54 Mostra d'Oltremare Pad 20, Naples, 80125, ITALY
| | - Daniele D'Arrigo
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Marta Tomasini
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco chies 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Christian Candrian
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, via Tesserete 46, Lugano, 6900, SWITZERLAND
| | - Luigi Ambrosio
- Institute of Polymers Composites and Biomaterials National Research Council, Viale Kennedy, Pozzuoli, Campania, 80078, ITALY
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| |
Collapse
|
6
|
Soares MBP, Gonçalves RGJ, Vasques JF, da Silva-Junior AJ, Gubert F, Santos GC, de Santana TA, Almeida Sampaio GL, Silva DN, Dominici M, Mendez-Otero R. Current Status of Mesenchymal Stem/Stromal Cells for Treatment of Neurological Diseases. Front Mol Neurosci 2022; 15:883378. [PMID: 35782379 PMCID: PMC9244712 DOI: 10.3389/fnmol.2022.883378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurological disorders include a wide spectrum of clinical conditions affecting the central and peripheral nervous systems. For these conditions, which affect hundreds of millions of people worldwide, generally limited or no treatments are available, and cell-based therapies have been intensively investigated in preclinical and clinical studies. Among the available cell types, mesenchymal stem/stromal cells (MSCs) have been widely studied but as yet no cell-based treatment exists for neurological disease. We review current knowledge of the therapeutic potential of MSC-based therapies for neurological diseases, as well as possible mechanisms of action that may be explored to hasten the development of new and effective treatments. We also discuss the challenges for culture conditions, quality control, and the development of potency tests, aiming to generate more efficient cell therapy products for neurological disorders.
Collapse
Affiliation(s)
- Milena B. P. Soares
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Renata G. J. Gonçalves
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J. da Silva-Junior
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Gubert
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Girlaine Café Santos
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Thaís Alves de Santana
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Gabriela Louise Almeida Sampaio
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | | | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Rosalia Mendez-Otero
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Acevedo Rua L, Mumme M, Manferdini C, Darwiche S, Khalil A, Hilpert M, Buchner DA, Lisignoli G, Occhetta P, von Rechenberg B, Haug M, Schaefer DJ, Jakob M, Caplan A, Martin I, Barbero A, Pelttari K. Engineered nasal cartilage for the repair of osteoarthritic knee cartilage defects. Sci Transl Med 2021; 13:eaaz4499. [PMID: 34516821 DOI: 10.1126/scitranslmed.aaz4499] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lina Acevedo Rua
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcus Mumme
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Cristina Manferdini
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Salim Darwiche
- Musculoskeletal Research Unit MSRU, Equine Department, University of Zurich, 8057 Zürich, Switzerland
| | - Ahmad Khalil
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Morgane Hilpert
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Paola Occhetta
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Brigitte von Rechenberg
- Competence Center for Applied Biotechnology and Molecular Medicine CABMM, University of Zurich, 8057 Zürich, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Dirk J Schaefer
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcel Jakob
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Arnold Caplan
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14-16, 4123 Allschwil, Switzerland Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| |
Collapse
|
8
|
Aranda Hernandez J, Heuer C, Bahnemann J, Szita N. Microfluidic Devices as Process Development Tools for Cellular Therapy Manufacturing. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 179:101-127. [PMID: 34410457 DOI: 10.1007/10_2021_169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cellular therapies are creating a paradigm shift in the biomanufacturing industry. Particularly for autologous therapies, small-scale processing methods are better suited than the large-scale approaches that are traditionally employed in the industry. Current small-scale methods for manufacturing personalized cell therapies, however, are labour-intensive and involve a number of 'open events'. To overcome these challenges, new cell manufacturing platforms following a GMP-in-a-box concept have recently come on the market (GMP: Good Manufacturing Practice). These are closed automated systems with built-in pumps for fluid handling and sensors for in-process monitoring. At a much smaller scale, microfluidic devices exhibit many of the same features as current GMP-in-a-box systems. They are closed systems, fluids can be processed and manipulated, and sensors integrated for real-time detection of process variables. Fabricated from polymers, they can be made disposable, i.e. single-use. Furthermore, microfluidics offers exquisite spatiotemporal control over the cellular microenvironment, promising both reproducibility and control of outcomes. In this chapter, we consider the challenges in cell manufacturing, highlight recent advances of microfluidic devices for each of the main process steps, and summarize our findings on the current state of the art. As microfluidic cell culture devices have been reported for both adherent and suspension cell cultures, we report on devices for the key process steps, or unit operations, of both stem cell therapies and cell-based immunotherapies.
Collapse
Affiliation(s)
| | - Christopher Heuer
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Janina Bahnemann
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Nicolas Szita
- Biochemical Engineering Department, University College London (UCL), London, UK.
| |
Collapse
|
9
|
Soheilmoghaddam F, Rumble M, Cooper-White J. High-Throughput Routes to Biomaterials Discovery. Chem Rev 2021; 121:10792-10864. [PMID: 34213880 DOI: 10.1021/acs.chemrev.0c01026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many existing clinical treatments are limited in their ability to completely restore decreased or lost tissue and organ function, an unenviable situation only further exacerbated by a globally aging population. As a result, the demand for new medical interventions has increased substantially over the past 20 years, with the burgeoning fields of gene therapy, tissue engineering, and regenerative medicine showing promise to offer solutions for full repair or replacement of damaged or aging tissues. Success in these fields, however, inherently relies on biomaterials that are engendered with the ability to provide the necessary biological cues mimicking native extracellular matrixes that support cell fate. Accelerating the development of such "directive" biomaterials requires a shift in current design practices toward those that enable rapid synthesis and characterization of polymeric materials and the coupling of these processes with techniques that enable similarly rapid quantification and optimization of the interactions between these new material systems and target cells and tissues. This manuscript reviews recent advances in combinatorial and high-throughput (HT) technologies applied to polymeric biomaterial synthesis, fabrication, and chemical, physical, and biological screening with targeted end-point applications in the fields of gene therapy, tissue engineering, and regenerative medicine. Limitations of, and future opportunities for, the further application of these research tools and methodologies are also discussed.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Madeleine Rumble
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| |
Collapse
|
10
|
Jauković A, Abadjieva D, Trivanović D, Stoyanova E, Kostadinova M, Pashova S, Kestendjieva S, Kukolj T, Jeseta M, Kistanova E, Mourdjeva M. Specificity of 3D MSC Spheroids Microenvironment: Impact on MSC Behavior and Properties. Stem Cell Rev Rep 2021; 16:853-875. [PMID: 32681232 DOI: 10.1007/s12015-020-10006-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSC) have been considered the promising candidates for the regenerative and personalized medicine due to their self-renewal potential, multilineage differentiation and immunomodulatory capacity. Although these properties have encouraged profound MSC studies in recent years, the majority of research has been based on standard 2D culture utilization. The opportunity to resemble in vivo characteristics of cells native niche has been provided by implementation of 3D culturing models such as MSC spheroid formation assesed through cells self-assembling. In this review, we address the current literature on physical and biochemical features of 3D MSC spheroid microenvironment and their impact on MSC properties and behaviors. Starting with the reduction in the cells' dimensions and volume due to the changes in adhesion molecules expression and cytoskeletal proteins rearrangement resembling native conditions, through the microenvironment shifts in oxygen, nutrients and metabolites gradients and demands, we focus on distinctive and beneficial features of MSC in spheroids compared to cells cultured in 2D conditions. By summarizing the data for 3D MSC spheroids regarding cell survival, pluripotency, differentiation, immunomodulatory activities and potential to affect tumor cells growth we highlighted advantages and perspectives of MSC spheroids use in regenerative medicine. Further detailed analyses are needed to deepen our understanding of mechanisms responsible for modified MSC behavior in spheroids and to set future directions for MSC clinical application.
Collapse
Affiliation(s)
- Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia
| | - Desislava Abadjieva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia.,IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics, Röntgenring 11, D-97070, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Germany
| | - Elena Stoyanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Milena Kostadinova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Shina Pashova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Snejana Kestendjieva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Obilní trh 11, 602 00, Brno, Czech Republic.,Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Kamýcká 129, 165 00, Suchdol, Praha 6, Czech Republic
| | - Elena Kistanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Milena Mourdjeva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria.
| |
Collapse
|
11
|
Witte K, de Andrés MC, Wells J, Dalby MJ, Salmeron-Sanchez M, Oreffo ROC. Chondrobags: A high throughput alginate-fibronectin micromass platform for in vitro human cartilage formation. Biofabrication 2020; 12:045034. [DOI: 10.1088/1758-5090/abb653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
12
|
Afflerbach AK, Kiri MD, Detinis T, Maoz BM. Mesenchymal Stem Cells as a Promising Cell Source for Integration in Novel In Vitro Models. Biomolecules 2020; 10:E1306. [PMID: 32927777 PMCID: PMC7565384 DOI: 10.3390/biom10091306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
The human-relevance of an in vitro model is dependent on two main factors-(i) an appropriate human cell source and (ii) a modeling platform that recapitulates human in vivo conditions. Recent years have brought substantial advancements in both these aspects. In particular, mesenchymal stem cells (MSCs) have emerged as a promising cell source, as these cells can differentiate into multiple cell types, yet do not raise the ethical and practical concerns associated with other types of stem cells. In turn, advanced bioengineered in vitro models such as microfluidics, Organs-on-a-Chip, scaffolds, bioprinting and organoids are bringing researchers ever closer to mimicking complex in vivo environments, thereby overcoming some of the limitations of traditional 2D cell cultures. This review covers each of these advancements separately and discusses how the integration of MSCs into novel in vitro platforms may contribute enormously to clinical and fundamental research.
Collapse
Affiliation(s)
- Ann-Kristin Afflerbach
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Faculty of Biosciences, Universität Heidelberg, 69120 Heidelberg, Germany
| | - Mark D. Kiri
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Tahir Detinis
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
13
|
Volleman TNE, Schol J, Morita K, Sakai D, Watanabe M. Wnt3a and wnt5a as Potential Chondrogenic Stimulators for Nucleus Pulposus Cell Induction: A Comprehensive Review. Neurospine 2020; 17:19-35. [PMID: 32252152 PMCID: PMC7136098 DOI: 10.14245/ns.2040040.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/18/2020] [Indexed: 12/20/2022] Open
Abstract
Low back pain remains a highly prevalent pathology engendering a tremendous socioeconomic burden. Low back pain is generally associated with intervertebral disc (IVD) degeneration, a process involving the deterioration of nucleus pulpous (NP) cells and IVD matrix. Scientific interest has directed efforts to restoring cell numbers as a strategy to enable IVD regeneration. Currently, mesenchymal stromal cells (MSCs) are being explored as cell therapy agents, due to their easy accessibility and differentiation potential. For enhancement of MSCs, growth factor supplementation is commonly applied to induce differentiation towards a chondrogenic (NP) cell phenotype. The wnt signaling pathways play a crucial role in chondrogenesis, nonetheless, literature appears to present controversies with regard to wnt3a and wnt5a for the induction of NP cells, chondrocytes, and MSCs. This review aims to summarize the reporting on wnt3a/wnt5a mediated NP cell differentiation, and to elucidate the mechanisms involved in wnt3a and wnt5a mediated chondrogenesis for potential application as cell therapy supplements for IVD regeneration. Our review suggests that wnt3a, subsequently replaced with a chondrogenic stimulating growth factor, can enhance the chondrogenic potential of MSCs in vitro. Contrariwise, wnt5a is suggested to play a role in maintaining cell potency of differentiated NP or chondrogenic cells.
Collapse
Affiliation(s)
- Tibo Nico Emmie Volleman
- Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jordy Schol
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Kosuke Morita
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
14
|
da Silva Morais A, Vieira S, Zhao X, Mao Z, Gao C, Oliveira JM, Reis RL. Advanced Biomaterials and Processing Methods for Liver Regeneration: State-of-the-Art and Future Trends. Adv Healthc Mater 2020; 9:e1901435. [PMID: 31977159 DOI: 10.1002/adhm.201901435] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.
Collapse
Affiliation(s)
- Alain da Silva Morais
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Sílvia Vieira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Xinlian Zhao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Joaquim M. Oliveira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
15
|
Carvalho MR, Truckenmuller R, Reis RL, Oliveira JM. Biomaterials and Microfluidics for Drug Discovery and Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1230:121-135. [DOI: 10.1007/978-3-030-36588-2_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
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: 13] [Impact Index Per Article: 2.2] [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
|
17
|
Benmassaoud MM, Gultian KA, DiCerbo M, Vega SL. Hydrogel screening approaches for bone and cartilage tissue regeneration. Ann N Y Acad Sci 2019; 1460:25-42. [DOI: 10.1111/nyas.14247] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Matthew DiCerbo
- Department of Biomedical EngineeringRowan University Glassboro New Jersey
| | - Sebastián L. Vega
- Department of Biomedical EngineeringRowan University Glassboro New Jersey
| |
Collapse
|
18
|
Wang L, Jiang D, Wang Q, Wang Q, Hu H, Jia W. The Application of Microfluidic Techniques on Tissue Engineering in Orthopaedics. Curr Pharm Des 2019; 24:5397-5406. [PMID: 30827230 DOI: 10.2174/1381612825666190301142833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/20/2019] [Indexed: 12/15/2022]
Abstract
Background:
Tissue engineering (TE) is a promising solution for orthopaedic diseases such as bone or
cartilage defects and bone metastasis. Cell culture in vitro and scaffold fabrication are two main parts of TE, but
these two methods both have their own limitations. The static cell culture medium is unable to achieve multiple
cell incubation or offer an optimal microenvironment for cells, while regularly arranged structures are unavailable
in traditional cell-laden scaffolds, which results in low biocompatibility. To solve these problems, microfluidic
techniques are combined with TE. By providing 3-D networks and interstitial fluid flows, microfluidic platforms
manage to maintain phenotype and viability of osteocytic or chondrocytic cells, and the precise manipulation of
liquid, gel and air flows in microfluidic devices leads to the highly organized construction of scaffolds.
Methods:
In this review, we focus on the recent advances of microfluidic techniques applied in the field of tissue
engineering, especially in orthropaedics. An extensive literature search was done using PubMed. The introduction
describes the properties of microfluidics and how it exploits the advantages to the full in the aspects of TE. Then
we discuss the application of microfluidics on the cultivation of osteocytic cells and chondrocytes, and other
extended researches carried out on this platform. The following section focuses on the fabrication of highly organized
scaffolds and other biomaterials produced by microfluidic devices. Finally, the incubation and studying of
bone metastasis models in microfluidic platforms are discussed.
Conclusion:
The combination of microfluidics and tissue engineering shows great potentials in the osteocytic cell
culture and scaffold fabrication. Though there are several problems that still require further exploration, the future
of microfluidics in TE is promising.
Collapse
Affiliation(s)
- Lingtian Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Dajun Jiang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Qiyang Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Qing Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Haoran Hu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Weitao Jia
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| |
Collapse
|
19
|
Park K, Lee Y, Seo J. Recent Advances in High-throughput Platforms with Engineered Biomaterial Microarrays for Screening of Cell and Tissue Behavior. Curr Pharm Des 2019; 24:5458-5470. [DOI: 10.2174/1381612825666190207093438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 02/06/2023]
Abstract
In the last decades, bioengineers have developed myriad biomaterials for regenerative medicine. Development of screening techniques is essential for understanding complex behavior of cells in the biological microenvironments. Conventional approaches to the screening of cellular behavior in vitro have limitations in terms of accuracy, reusability, labor-intensive screening, and versatility. Thus, drug screening and toxicology test through in vitro screening platforms have been underwhelming. Recent advances in the high-throughput screening platforms somewhat overcome the limitations of in vitro screening platforms via repopulating human tissues’ biophysical and biomchemical microenvironments with the ability to continuous monitoring of miniaturized human tissue behavior. Herein, we review current trends in the screening platform in which a high-throughput system composed of engineered microarray devices is developed to investigate cell-biomaterial interaction. Furthermore, diverse methods to achieve continuous monitoring of cell behavior via developments of biosensor integrated high-throughput platforms, and future perspectives on high-throughput screening will be provided.
Collapse
Affiliation(s)
- Kijun Park
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Yeontaek Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Jungmok Seo
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| |
Collapse
|
20
|
Friddin MS, Elani Y, Trantidou T, Ces O. New Directions for Artificial Cells Using Prototyped Biosystems. Anal Chem 2019; 91:4921-4928. [PMID: 30841694 DOI: 10.1021/acs.analchem.8b04885] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Microfluidics has has enabled the generation of a range of single compartment and multicompartment vesicles and bilayer-delineated droplets that can be assembled in 2D and 3D. These model systems are becoming increasingly used as artificial cell chassis and as biomimetic constructs for assembling tissue models, engineering therapeutic delivery systems, and screening drugs. One bottleneck in developing this technology is the time, expertise, and equipment required for device fabrication. This has led to interest across the microfluidics community in using rapid prototyping to engineer microfluidic devices from computer-aided-design (CAD) drawings. We highlight how this rapid-prototyping revolution is transforming the fabrication of microfluidic devices for artificial cell construction in bottom-up synthetic biology. We provide an outline of the current landscape and present how advances in the field may give rise to the next generation of multifunctional biodevices, particularly with Industry 4.0 on the horizon. Successfully developing this technology and making it open-source could pave the way for a new generation of citizen-led science, fueling the possibility that the next multibillion-dollar start-up could emerge from an attic or a basement.
Collapse
Affiliation(s)
- Mark S Friddin
- Department of Chemistry , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom
| | - Yuval Elani
- Department of Chemistry , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom.,Institute of Chemical Biology , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom.,fabriCELL, Molecular Sciences Research Hub , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom
| | - Tatiana Trantidou
- Department of Chemistry , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom
| | - Oscar Ces
- Department of Chemistry , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom.,Institute of Chemical Biology , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom.,fabriCELL, Molecular Sciences Research Hub , Imperial College London , Wood Lane , London , W12 0BZ , United Kingdom
| |
Collapse
|
21
|
Vasilakis N, Papadimitriou KI, Morgan H, Prodromakis T. Modular Pressure and Flow Rate-Balanced Microfluidic Serial Dilution Networks for Miniaturised Point-of-Care Diagnostic Platforms. SENSORS 2019; 19:s19040911. [PMID: 30795601 PMCID: PMC6412972 DOI: 10.3390/s19040911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/28/2022]
Abstract
Fast, efficient and more importantly accurate serial dilution is a necessary requirement for most biochemical microfluidic-based quantitative diagnostic applications. Over the last two decades, a multitude of microfluidic devices has been proposed, each one demonstrating either a different type of dilution technique or complex system architecture based on various flow source and valving combinations. In this work, a novel serial dilution network architecture is demonstrated, implemented on two entirely different substrates for validation and performance characterisation. The single layer, stepwise serial diluter comprises an optimised microfluidic network, where identical dilution ratios per stage are ensured, either by applying equal pressure or equal flow rates at both inlets. The advantages of this serial diluter are twofold: Firstly, it is structured as a modular unit cell, simplifying the required fluid driving mechanism to a single source for both sample and buffer solution. Thus, this unit cell can be used as a fundamental microfluidic building block, forming multistage serial dilution cascades, once combined appropriately with itself or other similar unit cells. Secondly, the serial diluter can tolerate the inevitable flow source fluctuations, ensuring constant dilution ratios without the need to employ damping mechanisms, making it ideal for Point of Care (PoC) platforms. Proof-of-concept experiments with glucose have demonstrated good agreement between simulations and measurements, highlighting the validity of our serial diluter.
Collapse
Affiliation(s)
- Nikolaos Vasilakis
- Nanoelectronics & Nanotechnology Research Group, Electronics and Computer Science, University of Southampton, Southampton SO17 1BJ, UK.
| | - Konstantinos I Papadimitriou
- Nanoelectronics & Nanotechnology Research Group, Electronics and Computer Science, University of Southampton, Southampton SO17 1BJ, UK.
| | - Hywel Morgan
- Nanoelectronics & Nanotechnology Research Group, Electronics and Computer Science, University of Southampton, Southampton SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Themistoklis Prodromakis
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
22
|
Mahapatra C, Kim JJ, Lee JH, Jin GZ, Knowles JC, Kim HW. Differential chondro- and osteo-stimulation in three-dimensional porous scaffolds with different topological surfaces provides a design strategy for biphasic osteochondral engineering. J Tissue Eng 2019; 10:2041731419826433. [PMID: 30728938 PMCID: PMC6357292 DOI: 10.1177/2041731419826433] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/07/2019] [Indexed: 11/17/2022] Open
Abstract
Bone/cartilage interfacial tissue engineering needs to satisfy the differential properties and architectures of the osteochondral region. Therefore, biphasic or multiphasic scaffolds that aim to mimic the gradient hierarchy are widely used. Here, we find that two differently structured (topographically) three-dimensional scaffolds, namely, "dense" and "nanofibrous" surfaces, show differential stimulation in osteo- and chondro-responses of cells. While the nanofibrous scaffolds accelerate the osteogenesis of mesenchymal stem cells, the dense scaffolds are better in preserving the phenotypes of chondrocytes. Two types of porous scaffolds, generated by a salt-leaching method combined with a phase-separation process using the poly(lactic acid) composition, had a similar level of porosity (~90%) and pore size (~150 μm). The major difference in the surface nanostructure led to substantial changes in the surface area and water hydrophilicity (nanofibrous ≫ dense); as a result, the nanofibrous scaffolds increased the cell-to-matrix adhesion of mesenchymal stem cells significantly while decreasing the cell-to-cell contracts. Importantly, the chondrocytes, when cultured on nanofibrous scaffolds, were prone to lose their phenotype, including reduced chondrogenic expressions (SOX-9, collagen type II, and Aggrecan) and glycosaminoglycan content, which was ascribed to the enhanced cell-matrix adhesion with reduced cell-cell contacts. On the contrary, the osteogenesis of mesenchymal stem cells was significantly accelerated by the improved cell-to-matrix adhesion, as evidenced in the enhanced osteogenic expressions (RUNX2, bone sialoprotein, and osteopontin) and cellular mineralization. Based on these findings, we consider that the dense scaffold is preferentially used for the chondral-part, whereas the nanofibrous structure is suitable for osteo-part, to provide an optimal biphasic matrix environment for osteochondral tissue engineering.
Collapse
Affiliation(s)
- Chinmaya Mahapatra
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Jung-Ju Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Jonathan C Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
23
|
A Microfluidic Spheroid Culture Device with a Concentration Gradient Generator for High-Throughput Screening of Drug Efficacy. Molecules 2018; 23:molecules23123355. [PMID: 30567363 PMCID: PMC6321514 DOI: 10.3390/molecules23123355] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022] Open
Abstract
Three-dimensional (3D) cell culture is considered more clinically relevant in mimicking the structural and physiological conditions of tumors in vivo compared to two-dimensional cell cultures. In recent years, high-throughput screening (HTS) in 3D cell arrays has been extensively used for drug discovery because of its usability and applicability. Herein, we developed a microfluidic spheroid culture device (μFSCD) with a concentration gradient generator (CGG) that enabled cells to form spheroids and grow in the presence of cancer drug gradients. The device is composed of concave microwells with several serpentine micro-channels which generate a concentration gradient. Once the colon cancer cells (HCT116) formed a single spheroid (approximately 120 μm in diameter) in each microwell, spheroids were perfused in the presence of the cancer drug gradient irinotecan for three days. The number of spheroids, roundness, and cell viability, were inversely proportional to the drug concentration. These results suggest that the μFSCD with a CGG has the potential to become an HTS platform for screening the efficacy of cancer drugs.
Collapse
|
24
|
Duru LN, Quan Z, Qazi TJ, Qing H. Stem cells technology: a powerful tool behind new brain treatments. Drug Deliv Transl Res 2018; 8:1564-1591. [PMID: 29916013 DOI: 10.1007/s13346-018-0548-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cell research has recently become a hot research topic in biomedical research due to the foreseen unlimited potential of stem cells in tissue engineering and regenerative medicine. For many years, medicine has been facing intense challenges, such as an insufficient number of organ donations that is preventing clinicians to fulfill the increasing needs. To try and overcome this regrettable matter, research has been aiming at developing strategies to facilitate the in vitro culture and study of stem cells as a tool for tissue regeneration. Meanwhile, new developments in the microfluidics technology brought forward emerging cell culture applications that are currently allowing for a better chemical and physical control of cellular microenvironment. This review presents the latest developments in stem cell research that brought new therapies to the clinics and how the convergence of the microfluidics technology with stem cell research can have positive outcomes on the fields of regenerative medicine and high-throughput screening. These advances will bring new translational solutions for drug discovery and will upgrade in vitro cell culture to a new level of accuracy and performance. We hope this review will provide new insights into the understanding of new brain treatments from the perspective of stem cell technology especially regarding regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Lucienne N Duru
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhenzhen Quan
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Talal Jamil Qazi
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing, China. .,Beijing Key Laboratory of Separation and Analysis in Biomedical and Pharmaceuticals, Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Haidian District, Beijing, 100081, China.
| |
Collapse
|
25
|
Lembong J, Lerman MJ, Kingsbury TJ, Civin CI, Fisher JP. A Fluidic Culture Platform for Spatially Patterned Cell Growth, Differentiation, and Cocultures. Tissue Eng Part A 2018; 24:1715-1732. [PMID: 29845891 PMCID: PMC6302678 DOI: 10.1089/ten.tea.2018.0020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/24/2018] [Indexed: 01/09/2023] Open
Abstract
Stem cell cultures within perfusion bioreactors, while efficient in obtaining cell numbers, often lack the similarity to native tissues and consequently cell phenotype. We develop a three-dimensional (3D)-printed fluidic chamber for dynamic stem cell culture, with emphasis on control over flow and substrate curvature in a 3D environment, two physiologic features of native tissues. The chamber geometry, consisting of an array of vertical cylindrical pillars, facilitates actin-mediated localization of human mesenchymal stem cells (hMSCs) within ∼200 μm distance from the pillars, enabling spatial patterning of hMSCs and endothelial cells in cocultures and subsequent modulation of calcium signaling between these two essential cell types in the bone marrow microenvironment. Flow-enhanced osteogenic differentiation of hMSCs in growth media imposes spatial variations of alkaline phosphatase expression, which positively correlates with local shear stress. Proliferation of hMSCs is maintained within the chamber, exceeding the cell expansion in conventional static culture. The capability to manipulate cell spatial patterning, differentiation, and 3D tissue formation through geometry and flow demonstrates the culture chamber's relevant chemomechanical cues in stem cell microenvironments, thus providing an easy-to-implement tool to study interactions among substrate curvature, shear stress, and intracellular actin machinery in the tissue-engineered construct.
Collapse
Affiliation(s)
- Josephine Lembong
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
- NIH Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Max J. Lerman
- NIH Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Tami J. Kingsbury
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Curt I. Civin
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
- NIH Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| |
Collapse
|
26
|
Abstract
Microfluidics has played a vital role in developing novel methods to investigate biological phenomena at the molecular and cellular level during the last two decades. Microscale engineering of cellular systems is nevertheless a nascent field marked inherently by frequent disruptive advancements in technology such as PDMS-based soft lithography. Viable culture and manipulation of cells in microfluidic devices requires knowledge across multiple disciplines including molecular and cellular biology, chemistry, physics, and engineering. There has been numerous excellent reviews in the past 15 years on applications of microfluidics for molecular and cellular biology including microfluidic cell culture (Berthier et al., 2012; El-Ali, Sorger, & Jensen, 2006; Halldorsson et al., 2015; Kim et al., 2007; Mehling & Tay, 2014; Sackmann et al., 2014; Whitesides, 2006; Young & Beebe, 2010), cell culture models (Gupta et al., 2016; Inamdar & Borenstein, 2011; Meyvantsson & Beebe, 2008), cell secretion (Schrell et al., 2016), chemotaxis (Kim & Wu, 2012; Wu et al., 2013), neuron culture (Millet & Gillette, 2012a, 2012b), drug screening (Dittrich & Manz, 2006; Eribol, Uguz, & Ulgen, 2016; Wu, Huang, & Lee, 2010), cell sorting (Autebert et al., 2012; Bhagat et al., 2010; Gossett et al., 2010; Wyatt Shields Iv, Reyes, & López, 2015), single cell studies (Lecault et al., 2012; Reece et al., 2016; Yin & Marshall, 2012), stem cell biology (Burdick & Vunjak-Novakovic, 2009; Wu et al., 2011; Zhang & Austin, 2012), cell differentiation (Zhang et al., 2017a), systems biology (Breslauer, Lee, & Lee, 2006), 3D cell culture (Huh et al., 2011; Li et al., 2012; van Duinen et al., 2015), spheroids and organoids (Lee et al., 2016; Montanez-Sauri, Beebe, & Sung, 2015; Morimoto & Takeuchi, 2013; Skardal et al., 2016; Young, 2013), organ-on-chip (Bhatia & Ingber, 2014; Esch, Bahinski, & Huh, 2015; Huh et al., 2011; van der Meer & van den Berg, 2012), and tissue engineering (Andersson & Van Den Berg, 2004; Choi et al., 2007; Hasan et al., 2014). In this chapter, we provide an overview of PDMS-based microdevices for microfluidic cell culture. We discuss the advantages and challenges of using PDMS-based soft lithography for microfluidic cell culture and highlight recent progress and future directions in this area.
Collapse
Affiliation(s)
- Melikhan Tanyeri
- Biomedical Engineering Program, Duquesne University, Pittsburgh, PA, United States
| | - Savaş Tay
- Institute of Molecular Engineering, University of Chicago, Chicago, IL, United States; Institute of Genomics and Systems Biology, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
27
|
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]
|
28
|
Ugolini GS, Visone R, Cruz-Moreira D, Mainardi A, Rasponi M. Generation of functional cardiac microtissues in a beating heart-on-a-chip. Methods Cell Biol 2018; 146:69-84. [PMID: 30037467 DOI: 10.1016/bs.mcb.2018.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the increasing attention on cardiovascular disorders and the current inability of pre-clinical models to accurately predict human physiology, the need for advanced and reliable heart in vitro models is paramount. Microfabrication technologies provide potential solutions in the organs-on-chip systems: microengineered devices where cell cultures can be hosted and cultured to develop three-dimensional models or microtissues with high similarity to human physiology. We here described the fabrication and operation procedures for a beating heart-on-a-chip. The device features a culture region for a 3D cardiac microtissue and a system for applying tuned mechanical stimulation during culture to improve cardiac development. We additionally describe procedures for characterizing tissue maturation via immunofluorescence and functional evaluations of microtissue contractility.
Collapse
Affiliation(s)
| | - Roberta Visone
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Daniela Cruz-Moreira
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Andrea Mainardi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy.
| |
Collapse
|
29
|
Park MJ, Lee J, Byeon JS, Jeong DU, Gu NY, Cho IS, Cha SH. Effects of three-dimensional spheroid culture on equine mesenchymal stem cell plasticity. Vet Res Commun 2018; 42:171-181. [PMID: 29721754 DOI: 10.1007/s11259-018-9720-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 04/02/2018] [Indexed: 01/01/2023]
Abstract
Mesenchymal stem cells (MSCs) are useful candidates for tissue engineering and cell therapy fields. We optimize culture conditions of equine adipose tissue-derived MSCs (eAD-MSCs) for treatment of horse fractures. To investigate enhancing properties of three-dimensional (3D) culture system in eAD-MSCs, we performed various sized spheroid formation and determined changes in gene expression levels to obtain different sized spheroid for cell therapy. eAD-MSCs were successfully isolated from horse tailhead. Using hanging drop method, spheroid formation was generated for three days. Quantitative real-time PCR was performed to analyze gene expression. As results, expression levels of pluripotent markers were increased depending on spheroid size and the production of PGE2 was increased in spheroid formation compared to that in monolayer. Ki-67 showed a remarkable increase in the spheroid formed with 2.0 × 105 cells/drop as compared to that in the monolayer. Expression levels of angiogenesis-inducing factors such as VEGF, IL-6, IL-8, and IL-18 were significantly increased in spheroid formation compared to those in the monolayer. Expression levels of bone morphogenesis-inducing factors such as Cox-2 and TGF-β1 were also significantly increased in spheroid formation compared to those in the monolayer. Expression levels of osteocyte-specific markers such as RUNX2, osteocalcin, and differentiation potential were also significantly increased in spheroid formation compared to those in the monolayer. Therefore, spheroid formation of eAD-MSCs through the hanging drop method can increases the expression of angiogenesis-inducing and bone morphogenesis-inducing factors under optimal culture conditions.
Collapse
Affiliation(s)
- Mi Jeong Park
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jeong Su Byeon
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Da-Un Jeong
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - In-Soo Cho
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Sang-Ho Cha
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea.
| |
Collapse
|
30
|
Developmentally inspired programming of adult human mesenchymal stromal cells toward stable chondrogenesis. Proc Natl Acad Sci U S A 2018; 115:4625-4630. [PMID: 29666250 DOI: 10.1073/pnas.1720658115] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is generally accepted that adult human bone marrow-derived mesenchymal stromal cells (hMSCs) are default committed toward osteogenesis. Even when induced to chondrogenesis, hMSCs typically form hypertrophic cartilage that undergoes endochondral ossification. Because embryonic mesenchyme is obviously competent to generate phenotypically stable cartilage, it is questioned whether there is a correspondence between mesenchymal progenitor compartments during development and in adulthood. Here we tested whether forcing specific early events of articular cartilage development can program hMSC fate toward stable chondrogenesis. Inspired by recent findings that spatial restriction of bone morphogenetic protein (BMP) signaling guides embryonic progenitors toward articular cartilage formation, we hypothesized that selective inhibition of BMP drives the phenotypic stability of hMSC-derived chondrocytes. Two BMP type I receptor-biased kinase inhibitors were screened in a microfluidic platform for their time- and dose-dependent effect on hMSC chondrogenesis. The different receptor selectivity profile of tested compounds allowed demonstration that transient blockade of both ALK2 and ALK3 receptors, while permissive to hMSC cartilage formation, is necessary and sufficient to maintain a stable chondrocyte phenotype. Remarkably, even upon compound removal, hMSCs were no longer competent to undergo hypertrophy in vitro and endochondral ossification in vivo, indicating the onset of a constitutive change. Our findings demonstrate that adult hMSCs effectively share properties of embryonic mesenchyme in the formation of transient but also of stable cartilage. This opens potential pharmacological strategies to articular cartilage regeneration and more broadly indicates the relevance of developmentally inspired protocols to control the fate of adult progenitor cell systems.
Collapse
|
31
|
Super-resolution for asymmetric resolution of FIB-SEM 3D imaging using AI with deep learning. Sci Rep 2018; 8:5877. [PMID: 29651011 PMCID: PMC5897388 DOI: 10.1038/s41598-018-24330-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/23/2018] [Indexed: 11/15/2022] Open
Abstract
Scanning electron microscopy equipped with a focused ion beam (FIB-SEM) is a promising three-dimensional (3D) imaging technique for nano- and meso-scale morphologies. In FIB-SEM, the specimen surface is stripped by an ion beam and imaged by an SEM installed orthogonally to the FIB. The lateral resolution is governed by the SEM, while the depth resolution, i.e., the FIB milling direction, is determined by the thickness of the stripped thin layer. In most cases, the lateral resolution is superior to the depth resolution; hence, asymmetric resolution is generated in the 3D image. Here, we propose a new approach based on an image-processing or deep-learning-based method for super-resolution of 3D images with such asymmetric resolution, so as to restore the depth resolution to achieve symmetric resolution. The deep-learning-based method learns from high-resolution sub-images obtained via SEM and recovers low-resolution sub-images parallel to the FIB milling direction. The 3D morphologies of polymeric nano-composites are used as test images, which are subjected to the deep-learning-based method as well as conventional methods. We find that the former yields superior restoration, particularly as the asymmetric resolution is increased. Our super-resolution approach for images having asymmetric resolution enables observation time reduction.
Collapse
|
32
|
Acevedo JP, Angelopoulos I, van Noort D, Khoury M. Microtechnology applied to stem cells research and development. Regen Med 2018; 13:233-248. [PMID: 29557299 DOI: 10.2217/rme-2017-0123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Microfabrication and microfluidics contribute to the research of cellular functions of cells and their interaction with their environment. Previously, it has been shown that microfluidics can contribute to the isolation, selection, characterization and migration of cells. This review aims to provide stem cell researchers with a toolkit of microtechnology (mT) instruments for elucidating complex stem cells functions which are challenging to decipher with traditional assays and animal models. These microdevices are able to investigate about the differentiation and niche interaction, stem cells transcriptomics, therapeutic functions and the capture of their secreted microvesicles. In conclusion, microtechnology will allow a more realistic assessment of stem cells properties, driving and accelerating the translation of regenerative medicine approaches to the clinic.
Collapse
Affiliation(s)
- Juan Pablo Acevedo
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile
| | - Ioannis Angelopoulos
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile
| | - Danny van Noort
- Facultad de Ingeniería y Ciencias Aplicadas Universidad de los Andes, Santiago, Chile.,Biotechnology, IFM, Linköping University, Sweden
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Santiago, Chile
| |
Collapse
|
33
|
Zhang J, Yun S, Bi J, Dai S, Du Y, Zannettino ACW, Zhang H. Enhanced multi-lineage differentiation of human mesenchymal stem/stromal cells within poly(N-isopropylacrylamide-acrylic acid) microgel-formed three-dimensional constructs. J Mater Chem B 2018; 6:1799-1814. [PMID: 32254252 DOI: 10.1039/c8tb00376a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human mesenchymal stem/stromal cells (hMSCs) are a potential cell source of stem cell therapy for many serious diseases and hMSC spheroids have emerged to replace single cell suspensions for cell therapy. Three-dimensional (3D) scaffolds or hydrogels which can mimic properties of the extracellular matrix (ECM) have been widely explored for their application in tissue regeneration. However, there are considerably less studies on inducing differentiation of hMSC spheroids using 3D scaffolds or hydrogels. This study is the first to explore multi-lineage differentiation of a stem cell line and primary stem cells within poly(N-isopropylacrylamide) (p(NIPAAm))-based thermosensitive microgel-formed constructs. We first demonstrated that poly(N-isopropylacrylamide-co-acrylic acid) (p(NIPAAm-AA)) was not toxic to hMSCs and the microgel-formed constructs facilitated formation of uniform stem cell spheroids. Due to functional enhancement of cell spheroids, hMSCs within the 3D microgel-formed constructs were induced for multi-lineage differentiation as evidenced by significant up-regulation of messenger RNA (mRNA) expression of chondrogenic and osteogenic genes even in the absence of induction media on day 9. When induction media were in situ supplied on day 9, mRNA expression of chondrogenic, osteogenic and adipogenic genes within the microgel-formed constructs were significantly higher than that in the pellet and 2D cultures, respectively, on day 37. In addition, histological and immunofluorescent images also confirmed successful multi-lineage differentiation of hMSCs within the 3D microgel-formed constructs. Hence, the thermosensitive p(NIPAAm-AA) microgel can be potentially used in an in vitro model for cell differentiation or in vivo transplantation of pre-differentiated human mesenchymal stromal cells into patients for specific lineage differentiation.
Collapse
Affiliation(s)
- Jiabin Zhang
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | | | | | | | | | | | | |
Collapse
|
34
|
Lopa S, Mondadori C, Mainardi VL, Talò G, Costantini M, Candrian C, Święszkowski W, Moretti M. Translational Application of Microfluidics and Bioprinting for Stem Cell-Based Cartilage Repair. Stem Cells Int 2018; 2018:6594841. [PMID: 29535776 PMCID: PMC5838503 DOI: 10.1155/2018/6594841] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/06/2017] [Accepted: 12/05/2017] [Indexed: 01/09/2023] Open
Abstract
Cartilage defects can impair the most elementary daily activities and, if not properly treated, can lead to the complete loss of articular function. The limitations of standard treatments for cartilage repair have triggered the development of stem cell-based therapies. In this scenario, the development of efficient cell differentiation protocols and the design of proper biomaterial-based supports to deliver cells to the injury site need to be addressed through basic and applied research to fully exploit the potential of stem cells. Here, we discuss the use of microfluidics and bioprinting approaches for the translation of stem cell-based therapy for cartilage repair in clinics. In particular, we will focus on the optimization of hydrogel-based materials to mimic the articular cartilage triggered by their use as bioinks in 3D bioprinting applications, on the screening of biochemical and biophysical factors through microfluidic devices to enhance stem cell chondrogenesis, and on the use of microfluidic technology to generate implantable constructs with a complex geometry. Finally, we will describe some new bioprinting applications that pave the way to the clinical use of stem cell-based therapies, such as scaffold-free bioprinting and the development of a 3D handheld device for the in situ repair of cartilage defects.
Collapse
Affiliation(s)
- Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Carlotta Mondadori
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Valerio Luca Mainardi
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
- Laboratory of Biological Structures Mechanics-Chemistry, Material and Chemical Engineering Department “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Marco Costantini
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Christian Candrian
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
- Unità di Traumatologia e Ortopedia-ORL, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Wojciech Święszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
- Swiss Institute for Regenerative Medicine, Lugano, Switzerland
| |
Collapse
|
35
|
Marklein RA, Lam J, Guvendiren M, Sung KE, Bauer SR. Functionally-Relevant Morphological Profiling: A Tool to Assess Cellular Heterogeneity. Trends Biotechnol 2018; 36:105-118. [DOI: 10.1016/j.tibtech.2017.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/11/2017] [Accepted: 10/18/2017] [Indexed: 12/16/2022]
|
36
|
Seo J, Shin JY, Leijten J, Jeon O, Camci-Unal G, Dikina AD, Brinegar K, Ghaemmaghami AM, Alsberg E, Khademhosseini A. High-throughput approaches for screening and analysis of cell behaviors. Biomaterials 2018; 153:85-101. [PMID: 29079207 PMCID: PMC5702937 DOI: 10.1016/j.biomaterials.2017.06.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023]
Abstract
The rapid development of new biomaterials and techniques to modify them challenge our capability to characterize them using conventional methods. In response, numerous high-throughput (HT) strategies are being developed to analyze biomaterials and their interactions with cells using combinatorial approaches. Moreover, these systematic analyses have the power to uncover effects of delivered soluble bioactive molecules on cell responses. In this review, we describe the recent developments in HT approaches that help identify cellular microenvironments affecting cell behaviors and highlight HT screening of biochemical libraries for gene delivery, drug discovery, and toxicological studies. We also discuss HT techniques for the analyses of cell secreted biomolecules and provide perspectives on the future utility of HT approaches in biomedical engineering.
Collapse
Affiliation(s)
- Jungmok Seo
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Center for Biomaterials, Korea Institute of Science and Technology, 14 Hwarang-ro, Seongbuk-gu, Seoul, 02792, South Korea
| | - Jung-Youn Shin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Gulden Camci-Unal
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Department of Chemical Engineering, University of Massachusetts Lowell, 1 University Ave, Lowell, MA, 01854-2827, USA
| | - Anna D Dikina
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Katelyn Brinegar
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH, 44106, USA; National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul, 143-701, Republic of Korea; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA; Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia.
| |
Collapse
|
37
|
Lam J, Lee EJ, Clark EC, Mikos AG. Honing Cell and Tissue Culture Conditions for Bone and Cartilage Tissue Engineering. Cold Spring Harb Perspect Med 2017; 7:a025734. [PMID: 28348176 PMCID: PMC5710100 DOI: 10.1101/cshperspect.a025734] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
An avenue of tremendous interest and need in health care encompasses the regeneration of bone and cartilage. Over the years, numerous tissue engineering strategies have contributed substantial progress toward the realization of clinically relevant therapies. Cell and tissue culture protocols, however, show many variations that make experimental results among different publications challenging to compare. This collection surveys prevalent cell sources, soluble factors, culture medium formulations, environmental factors, and genetic modification approaches in the literature. The intent of consolidating this information is to provide a starting resource for scientists considering how to optimize the parameters for cell differentiation and tissue culture procedures within the context of bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- Johnny Lam
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Esther J Lee
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Elisa C Clark
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Antonios G Mikos
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77251
| |
Collapse
|
38
|
Mendes LF, Tam WL, Chai YC, Geris L, Luyten FP, Roberts SJ. Combinatorial Analysis of Growth Factors Reveals the Contribution of Bone Morphogenetic Proteins to Chondrogenic Differentiation of Human Periosteal Cells. Tissue Eng Part C Methods 2017; 22:473-86. [PMID: 27018617 DOI: 10.1089/ten.tec.2015.0436] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Successful application of cell-based strategies in cartilage and bone tissue engineering has been hampered by the lack of robust protocols to efficiently differentiate mesenchymal stem cells into the chondrogenic lineage. The development of chemically defined culture media supplemented with growth factors (GFs) has been proposed as a way to overcome this limitation. In this work, we applied a fractional design of experiment (DoE) strategy to screen the effect of multiple GFs (BMP2, BMP6, GDF5, TGF-β1, and FGF2) on chondrogenic differentiation of human periosteum-derived mesenchymal stem cells (hPDCs) in vitro. In a micromass culture (μMass) system, BMP2 had a positive effect on glycosaminoglycan deposition at day 7 (p < 0.001), which in combination with BMP6 synergistically enhanced cartilage-like tissue formation that displayed in vitro mineralization capacity at day 14 (p < 0.001). Gene expression of μMasses cultured for 7 days with a medium formulation supplemented with 100 ng/mL of BMP2 and BMP6 and a low concentration of GDF5, TGF-β1, and FGF2 showed increased expression of Sox9 (1.7-fold) and the matrix molecules aggrecan (7-fold increase) and COL2A1 (40-fold increase) compared to nonstimulated control μMasses. The DoE analysis indicated that in GF combinations, BMP2 was the strongest effector for chondrogenic differentiation of hPDCs. When transplanted ectopically in nude mice, the in vitro-differentiated μMasses showed maintenance of the cartilaginous phenotype after 4 weeks in vivo. This study indicates the power of using the DoE approach for the creation of new medium formulations for skeletal tissue engineering approaches.
Collapse
Affiliation(s)
- Luis Filipe Mendes
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Wai Long Tam
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Yoke Chin Chai
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Liesbet Geris
- 2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium .,3 Biomechanics Research Unit, University of Liege , Liege, Belgium .,4 Department of Mechanical Engineering, Biomechanics Section, Katholieke Universiteit Leuven, Heverlee, Belgium
| | - Frank P Luyten
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Scott J Roberts
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium .,5 Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London , The Royal National Orthopaedic Hospital, London, United Kingdom
| |
Collapse
|
39
|
Marcucio RS, Qin L, Alsberg E, Boerckel JD. Reverse engineering development: Crosstalk opportunities between developmental biology and tissue engineering. J Orthop Res 2017; 35:2356-2368. [PMID: 28660712 DOI: 10.1002/jor.23636] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
The fields of developmental biology and tissue engineering have been revolutionized in recent years by technological advancements, expanded understanding, and biomaterials design, leading to the emerging paradigm of "developmental" or "biomimetic" tissue engineering. While developmental biology and tissue engineering have long overlapping histories, the fields have largely diverged in recent years at the same time that crosstalk opportunities for mutual benefit are more salient than ever. In this perspective article, we will use musculoskeletal development and tissue engineering as a platform on which to discuss these emerging crosstalk opportunities and will present our opinions on the bright future of these overlapping spheres of influence. The multicellular programs that control musculoskeletal development are rapidly becoming clarified, represented by shifting paradigms in our understanding of cellular function, identity, and lineage specification during development. Simultaneously, advancements in bioartificial matrices that replicate the biochemical, microstructural, and mechanical properties of developing tissues present new tools and approaches for recapitulating development in tissue engineering. Here, we introduce concepts and experimental approaches in musculoskeletal developmental biology and biomaterials design and discuss applications in tissue engineering as well as opportunities for tissue engineering approaches to inform our understanding of fundamental biology. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2356-2368, 2017.
Collapse
Affiliation(s)
- Ralph S Marcucio
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania
| | - Eben Alsberg
- Departments of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, Cleveland, Ohio
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania.,Department of Bioengineering, University of Pennslyvania, Philadelphia, Pennsylvania.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
40
|
Jackson-Holmes EL, McDevitt TC, Lu H. A microfluidic trap array for longitudinal monitoring and multi-modal phenotypic analysis of individual stem cell aggregates. LAB ON A CHIP 2017; 17:3634-3642. [PMID: 28952622 PMCID: PMC5656523 DOI: 10.1039/c7lc00763a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Three-dimensional pluripotent stem cell (PSC) cultures have the ability to undergo differentiation, self-organization, and morphogenesis to yield complex, in vitro tissue models that recapitulate key elements of native tissues. These tissue models offer a system for studying mechanisms of tissue development, investigating disease mechanisms, and performing drug screening. It remains challenging, however, to standardize PSC aggregate differentiation and morphogenesis methods due to heterogeneity stemming from biological and environmental sources. It is also difficult to monitor and assess large numbers of individual samples longitudinally throughout culture using typical batch-based culture methods. To address these challenges, we have developed a microfluidic platform for culture, longitudinal monitoring, and phenotypic analysis of individual stem cell aggregates. This platform uses a hydrodynamic loading principle to capture pre-formed stem cell aggregates in independent traps. We demonstrated that multi-day culture of aggregates in this platform reduces heterogeneity in phenotypic parameters such as size and morphology. Additionally, we showed that culture and analysis steps can be performed sequentially in the same platform, enabling correlation of multiple modes of analysis for individual samples. We anticipate this platform being applied to improve abilities for phenotypic analysis of PSC aggregate tissues and to facilitate research in standardizing culture systems in order to dually increase the yield and reduce the heterogeneity of PSC-derived tissues.
Collapse
Affiliation(s)
- E L Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| | | | | |
Collapse
|
41
|
Fernando WA, Papantoniou I, Mendes LF, Hall GN, Bosmans K, Tam WL, Teixeira LM, Moos M, Geris L, Luyten FP. Limb derived cells as a paradigm for engineering self-assembling skeletal tissues. J Tissue Eng Regen Med 2017; 12:794-807. [PMID: 28603948 DOI: 10.1002/term.2498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/21/2022]
Abstract
Mimicking developmental events has been proposed as a strategy to engineer tissue constructs for regenerative medicine. However, this approach has not yet been investigated for skeletal tissues. Here, it is demonstrated that ectopic implantation of day-14.5 mouse embryonic long bone anlagen, dissociated into single cells and randomly incorporated in a bioengineered construct, gives rise to epiphyseal growth plate-like structures, bone and marrow, which share many morphological and molecular similarities to epiphyseal units that form after transplanting intact long bone anlage, demonstrating substantial robustness and autonomy of complex tissue self-assembly and the overall organogenesis process. In vitro studies confirm the self-aggregation and patterning capacity of anlage cells and demonstrate that the model can be used to evaluate the effects of large and small molecules on biological behaviour. These results reveal the preservation of self-organizing and self-patterning capacity of anlage cells even when disconnected from their developmental niche and subjected to system perturbations such as cellular dissociation. These inherent features make long bone anlage cells attractive as a model system for tissue engineering technologies aimed at creating constructs that have the potential to self-assemble and self-pattern complex architectural structures.
Collapse
Affiliation(s)
- Warnakulasuriya A Fernando
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Ioannis Papantoniou
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Luis F Mendes
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Gabriella Nilsson Hall
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Kathleen Bosmans
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Wai L Tam
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Liliana M Teixeira
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Malcolm Moos
- Division of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Liesbet Geris
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium.,Biomechanics Research Unit, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| |
Collapse
|
42
|
Cha JM, Park H, Shin EK, Sung JH, Kim O, Jung W, Bang OY, Kim J. A novel cylindrical microwell featuring inverted-pyramidal opening for efficient cell spheroid formation without cell loss. Biofabrication 2017; 9:035006. [DOI: 10.1088/1758-5090/aa8111] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
43
|
McKee C, Chaudhry GR. Advances and challenges in stem cell culture. Colloids Surf B Biointerfaces 2017; 159:62-77. [PMID: 28780462 DOI: 10.1016/j.colsurfb.2017.07.051] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/22/2017] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) hold great promise for cell therapy, tissue engineering, and regenerative medicine as well as pharmaceutical and biotechnological applications. They have the capacity to self-renew and the ability to differentiate into specialized cell types depending upon their source of isolation. However, use of SCs for clinical applications requires a high quality and quantity of cells. This necessitates large-scale expansion of SCs followed by efficient and homogeneous differentiation into functional derivatives. Traditional methods for maintenance and expansion of cells rely on two-dimensional (2-D) culturing techniques using plastic culture plates and xenogenic media. These methods provide limited expansion and cells tend to lose clonal and differentiation capacity upon long-term passaging. Recently, new approaches for the expansion of SCs have emphasized three-dimensional (3-D) cell growth to mimic the in vivo environment. This review provides a comprehensive compendium of recent advancements in culturing SCs using 2-D and 3-D techniques involving spheroids, biomaterials, and bioreactors. In addition, potential challenges to achieve billion-fold expansion of cells are discussed.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
44
|
Kamperman T, Henke S, Zoetebier B, Ruiterkamp N, Wang R, Pouran B, Weinans H, Karperien M, Leijten J. Nanoemulsion-induced enzymatic crosslinking of tyramine-functionalized polymer droplets. J Mater Chem B 2017; 5:4835-4844. [PMID: 32263999 DOI: 10.1039/c7tb00686a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In situ gelation of water-in-oil polymer emulsions is a key method to produce hydrogel particles. Although this approach is in principle ideal for encapsulating bioactive components such as cells, the oil phase can interfere with straightforward presentation of crosslinker molecules. Several approaches have been developed to induce in-emulsion gelation by exploiting the triggered generation or release of crosslinker molecules. However, these methods typically rely on photo- or acid-based reactions that are detrimental to cell survival and functioning. In this work, we demonstrate the diffusion-based supplementation of small molecules for the in-emulsion gelation of multiple tyramine-functionalized polymers via enzymatic crosslinking using a H2O2/oil nanoemulsion. This strategy is compatible with various emulsification techniques, thereby readily supporting the formation of monodisperse hydrogel particles spanning multiple length scales ranging from the nano- to the millimeter. As proof of principle, we leveraged droplet microfluidics in combination with the cytocompatible nature of enzymatic crosslinking to engineer hollow cell-laden hydrogel microcapsules that support the formation of viable and functional 3D microtissues. The straightforward, universal, and cytocompatible nature of nanoemulsion-induced enzymatic crosslinking facilitates its rapid and widespread use in numerous food, pharma, and life science applications.
Collapse
Affiliation(s)
- Tom Kamperman
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, 7522NB Enschede, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Stem cell culture and differentiation in microfluidic devices toward organ-on-a-chip. Future Sci OA 2017; 3:FSO187. [PMID: 28670476 PMCID: PMC5481871 DOI: 10.4155/fsoa-2016-0091] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/21/2017] [Indexed: 02/07/2023] Open
Abstract
Microfluidic lab-on-a-chip provides a new platform with unique advantages to mimic complex physiological microenvironments in vivo and has been increasingly exploited to stem cell research. In this review, we highlight recent advances of microfluidic devices for stem cell culture and differentiation toward the development of organ-on-a-chip, especially with an emphasis on vital innovations within the last 2 years. Various aspects for improving on-chip stem-cell culture and differentiation, particularly toward organ-on-a-chip, are discussed, along with microenvironment control, surface modification, extracellular scaffolds, high throughput and stimuli. The combination of microfluidic technologies and stem cells hold great potential toward versatile systems of ‘organ-on-a-chip’ as desired.
Adapted with permission from [1–8]. Stem cells, capable of self-renewing and differentiating into cells of various tissue types, are drawing more and more attention for their enormous potential in many clinically associated applications that include drug screening, disease modeling and regenerative medicine. Conventional cell culture methods, however, have proven to be difficult to mimic in vivo like microenvironments and to provide a number of well-controlled stimuli that are critical for stem cell culture and differentiation. In contrast, microfluidic devices offer new capacities and unique advantages to mimic complex physiological microenvironments in vivo, and has been increasingly applied to stem cell research.
Collapse
|
46
|
Marti-Figueroa CR, Ashton RS. The case for applying tissue engineering methodologies to instruct human organoid morphogenesis. Acta Biomater 2017; 54:35-44. [PMID: 28315813 DOI: 10.1016/j.actbio.2017.03.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/24/2017] [Accepted: 03/14/2017] [Indexed: 12/20/2022]
Abstract
Three-dimensional organoids derived from human pluripotent stem cell (hPSC) derivatives have become widely used in vitro models for studying development and disease. Their ability to recapitulate facets of normal human development during in vitro morphogenesis produces tissue structures with unprecedented biomimicry. Current organoid derivation protocols primarily rely on spontaneous morphogenesis processes to occur within 3-D spherical cell aggregates with minimal to no exogenous control. This yields organoids containing microscale regions of biomimetic tissues, but at the macroscale (i.e. 100's of microns to millimeters), the organoids' morphology, cytoarchitecture, and cellular composition are non-biomimetic and variable. The current lack of control over in vitro organoid morphogenesis at the microscale induces aberrations at the macroscale, which impedes realization of the technology's potential to reproducibly form anatomically correct human tissue units that could serve as optimal human in vitro models and even transplants. Here, we review tissue engineering methodologies that could be used to develop powerful approaches for instructing multiscale, 3-D human organoid morphogenesis. Such technological mergers are critically needed to harness organoid morphogenesis as a tool for engineering functional human tissues with biomimetic anatomy and physiology. STATEMENT OF SIGNIFICANCE Human PSC-derived 3-D organoids are revolutionizing the biomedical sciences. They enable the study of development and disease within patient-specific genetic backgrounds and unprecedented biomimetic tissue microenvironments. However, their uncontrolled, spontaneous morphogenesis at the microscale yields inconsistences in macroscale organoid morphology, cytoarchitecture, and cellular composition that limits their standardization and application. Integration of tissue engineering methods with organoid derivation protocols could allow us to harness their potential by instructing standardized in vitro morphogenesis to generate organoids with biomimicry at all scales. Such advancements would enable the use of organoids as a basis for 'next-generation' tissue engineering of functional, anatomically mimetic human tissues and potentially novel organ transplants. Here, we discuss critical aspects of organoid morphogenesis where application of innovative tissue engineering methodologies would yield significant advancement towards this goal.
Collapse
|
47
|
Chimenti I, Massai D, Morbiducci U, Beltrami AP, Pesce M, Messina E. Stem Cell Spheroids and Ex Vivo Niche Modeling: Rationalization and Scaling-Up. J Cardiovasc Transl Res 2017; 10:150-166. [PMID: 28289983 DOI: 10.1007/s12265-017-9741-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
Abstract
Improved protocols/devices for in vitro culture of 3D cell spheroids may provide essential cues for proper growth and differentiation of stem/progenitor cells (S/PCs) in their niche, allowing preservation of specific features, such as multi-lineage potential and paracrine activity. Several platforms have been employed to replicate these conditions and to generate S/PC spheroids for therapeutic applications. However, they incompletely reproduce the niche environment, with partial loss of its highly regulated network, with additional hurdles in the field of cardiac biology, due to debated resident S/PCs therapeutic potential and clinical translation. In this contribution, the essential niche conditions (metabolic, geometric, mechanical) that allow S/PCs maintenance/commitment will be discussed. In particular, we will focus on both existing bioreactor-based platforms for the culture of S/PC as spheroids, and on possible criteria for the scaling-up of niche-like spheroids, which could be envisaged as promising tools for personalized cardiac regenerative medicine, as well as for high-throughput drug screening.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Rome, Italy
| | - Diana Massai
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino", IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry, "Umberto I" Hospital, "La Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
48
|
All-in-one 3D printed microscopy chamber for multidimensional imaging, the UniverSlide. Sci Rep 2017; 7:42378. [PMID: 28186188 PMCID: PMC5301227 DOI: 10.1038/srep42378] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022] Open
Abstract
While live 3D high resolution microscopy techniques are developing rapidly, their use for biological applications is partially hampered by practical difficulties such as the lack of a versatile sample chamber. Here, we propose the design of a multi-usage observation chamber adapted for live 3D bio-imaging. We show the usefulness and practicality of this chamber, which we named the UniverSlide, for live imaging of two case examples, namely multicellular systems encapsulated in sub-millimeter hydrogel shells and zebrafish larvae. We also demonstrate its versatility and compatibility with all microscopy devices by using upright or inverted microscope configurations after loading the UniverSlide with fixed or living samples. Further, the device is applicable for medium/high throughput screening and automatized multi-position image acquisition, providing a constraint-free but stable and parallelized immobilization of the samples. The frame of the UniverSlide is fabricated using a stereolithography 3D printer, has the size of a microscopy slide, is autoclavable and sealed with a removable lid, which makes it suitable for use in a controlled culture environment. We describe in details how to build this chamber and we provide all the files necessary to print the different pieces in the lab.
Collapse
|
49
|
|
50
|
Tsai AC, Liu Y, Yuan X, Chella R, Ma T. Aggregation kinetics of human mesenchymal stem cells under wave motion. Biotechnol J 2017; 12. [PMID: 27996210 DOI: 10.1002/biot.201600448] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/01/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are primary candidates in cell therapy and regenerative medicine but preserving their therapeutic potency following culture expansion is a significant challenge. hMSCs can spontaneously assemble into three-dimensional (3D) aggregates that enhance their regenerative properties. The present study investigated the impact of hydrodynamics conditions on hMSC aggregation kinetics under controlled rocking motion. While various laboratory methods have been developed for hMSC aggregate production, the rocking platform provides gentle mixing and can be scaled up using large bags as in wave motion bioreactors. The results show that the hMSC aggregation is mediated by cell adhesion molecules and that aggregate size distribution is influenced by seeding density, culture time, and hydrodynamic conditions. The analysis of fluid shear stress by COMSOL indicated that aggregate size distribution is inversely correlated with shear stress and that the rocking angle had a more pronounced effect on aggregate size distribution than the rocking speed due to its impact on shear stress. hMSC aggregates obtained from the bioreactor exhibit increased stemness, migratory properties, and expression of angiogenic factors. The results demonstrate the potential of the rocking platform to produce hMSC aggregates with controlled size distribution for therapeutic application.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Yijun Liu
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Ravindran Chella
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| |
Collapse
|