1
|
Han R, Luo L, Wei C, Qiao Y, Xie J, Pan X, Xing J. Stiffness-tunable biomaterials provide a good extracellular matrix environment for axon growth and regeneration. Neural Regen Res 2025; 20:1364-1376. [PMID: 39075897 PMCID: PMC11624885 DOI: 10.4103/nrr.nrr-d-23-01874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/31/2024] [Accepted: 03/16/2024] [Indexed: 07/31/2024] Open
Abstract
Neuronal growth, extension, branching, and formation of neural networks are markedly influenced by the extracellular matrix-a complex network composed of proteins and carbohydrates secreted by cells. In addition to providing physical support for cells, the extracellular matrix also conveys critical mechanical stiffness cues. During the development of the nervous system, extracellular matrix stiffness plays a central role in guiding neuronal growth, particularly in the context of axonal extension, which is crucial for the formation of neural networks. In neural tissue engineering, manipulation of biomaterial stiffness is a promising strategy to provide a permissive environment for the repair and regeneration of injured nervous tissue. Recent research has fine-tuned synthetic biomaterials to fabricate scaffolds that closely replicate the stiffness profiles observed in the nervous system. In this review, we highlight the molecular mechanisms by which extracellular matrix stiffness regulates axonal growth and regeneration. We highlight the progress made in the development of stiffness-tunable biomaterials to emulate in vivo extracellular matrix environments, with an emphasis on their application in neural repair and regeneration, along with a discussion of the current limitations and future prospects. The exploration and optimization of the stiffness-tunable biomaterials has the potential to markedly advance the development of neural tissue engineering.
Collapse
Affiliation(s)
- Ronglin Han
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Lanxin Luo
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Caiyan Wei
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yaru Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiming Xie
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xianchao Pan
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Juan Xing
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
2
|
Vanaei S, Hashemi M, Solouk A, Asghari Ilani M, Amili O, Hefzy MS, Tang Y, Elahinia M. Manufacturing, Processing, and Characterization of Self-Expanding Metallic Stents: A Comprehensive Review. Bioengineering (Basel) 2024; 11:983. [PMID: 39451359 PMCID: PMC11505524 DOI: 10.3390/bioengineering11100983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 10/26/2024] Open
Abstract
This paper aims to review the State of the Art in metal self-expanding stents made from nitinol (NiTi), showing shape memory and superelastic behaviors, to identify the challenges and the opportunities for improving patient outcomes. A significant contribution of this paper is its extensive coverage of multidisciplinary aspects, including design, simulation, materials development, manufacturing, bio/hemocompatibility, biomechanics, biomimicry, patency, and testing methodologies. Additionally, the paper offers in-depth insights into the latest practices and emerging trends, with a special emphasis on the transformative potential of additive manufacturing techniques in the development of metal stents. By consolidating existing knowledge and highlighting areas for future innovation, this review provides a valuable roadmap for advancing nitinol stents.
Collapse
Affiliation(s)
- Saeedeh Vanaei
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| | - Mahdi Hashemi
- Department of Materials and Metallurgical Engineering, Amirkabir University of Technology, Tehran 1591634311, Iran;
| | - Atefeh Solouk
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran;
| | - Mohsen Asghari Ilani
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran 1439957131, Iran;
| | - Omid Amili
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| | - Mohamed Samir Hefzy
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| | - Yuan Tang
- Department of Bioengineering, University of Toledo, Toledo, OH 43606, USA;
| | - Mohammad Elahinia
- Mechanical Industrial and Manufacturing Engineering Department, University of Toledo, Toledo, OH 43606, USA; (O.A.); (M.S.H.); (M.E.)
| |
Collapse
|
3
|
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
|
4
|
Jarrah R, Nathani KR, Bhandarkar S, Ezeudu CS, Nguyen RT, Amare A, Aljameey UA, Jarrah SI, Bhandarkar AR, Fiani B. Microfluidic 'brain-on chip' systems to supplement neurological practice: development, applications and considerations. Regen Med 2023; 18:413-423. [PMID: 37125510 DOI: 10.2217/rme-2022-0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Among the greatest general challenges in bioengineering is to mimic human physiology. Advanced efforts in tissue engineering have led to sophisticated 'brain-on-chip' (BoC) microfluidic devices that can mimic structural and functional aspects of brain tissue. BoC may be used to understand the biochemical pathways of neurolgical pathologies and assess promising therapeutic agents for facilitating regenerative medicine. We evaluated the potential of microfluidic BoC devices in various neurological pathologies, such as Alzheimer's, glioblastoma, traumatic brain injury, stroke and epilepsy. We also discuss the principles, limitations and future considerations of BoC technology. Results suggest that BoC models can help understand complex neurological pathologies and augment drug testing efforts for regenerative applications. However, implementing organ-on-chip technology to clinical practice has some practical limitations that warrant greater attention to improve large-scale applicability. Nevertheless, they remain to be versatile and powerful tools that can broaden our understanding of pathophysiological and therapeutic uncertainties to neurological diseases.
Collapse
Affiliation(s)
- Ryan Jarrah
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Shaan Bhandarkar
- Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Chibuze S Ezeudu
- Texas A&M School of Medicine,Texas A&M University, Bryan, TX 77807, USA
| | - Ryan T Nguyen
- University of Hawaii John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Abrham Amare
- Morehouse School of Medicine, Morehouse College, Atlanta, GA 30310, USA
| | - Usama A Aljameey
- Lincoln Memorial University DeBusk School of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Sabrina I Jarrah
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Brian Fiani
- Department of Neurosurgery, Cornell Medical Center/New York Presbyterian, New York, NY 10065, USA
| |
Collapse
|
5
|
Weber J, Weber M, Feile A, Schlensak C, Avci-Adali M. Development of an In Vitro Blood Vessel Model Using Autologous Endothelial Cells Generated from Footprint-Free hiPSCs to Analyze Interactions of the Endothelium with Blood Cell Components and Vascular Implants. Cells 2023; 12:cells12091217. [PMID: 37174617 PMCID: PMC10177426 DOI: 10.3390/cells12091217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Vascular implants, such as stents, are required to treat arterial stenosis or dilatation. The development of innovative stent materials and coatings, as well as novel preclinical testing strategies, is needed to improve the bio- and hemocompatibility of current stents. In this study, a blood vessel-like polydimethylsiloxane (PDMS) model was established to analyze the interaction of an endothelium with vascular implants, as well as blood-derived cells, in vitro. Using footprint-free human induced pluripotent stem cells (hiPSCs) and subsequent differentiation, functional endothelial cells (ECs) expressing specific markers were generated and used to endothelialize an artificial PDMS lumen. The established model was used to demonstrate the interaction of the created endothelium with blood-derived immune cells, which also allowed for real-time imaging. In addition, a stent was inserted into the endothelialized lumen to analyze the surface endothelialization of stents. In the future, this blood vessel-like model could serve as an in vitro platform to test the influence of vascular implants and coatings on endothelialization and to analyze the interaction of the endothelium with blood cell components.
Collapse
Affiliation(s)
- Josefin Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Marbod Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Adrian Feile
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| |
Collapse
|
6
|
Shin E, Kwon TY, Cho Y, Kim Y, Shin JH, Han YM. ECM Architecture-Mediated Regulation of β-Cell Differentiation from hESCs via Hippo-Independent YAP Activation. ACS Biomater Sci Eng 2023; 9:680-692. [PMID: 36580628 DOI: 10.1021/acsbiomaterials.2c01054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Changes in the extracellular matrix (ECM) influence stem cell fate. When hESCs were differentiated on a thin layer of Matrigel coated onto PDMS (Matrigel_PDMS), they exhibited a substantial increase in focal adhesion and focal adhesion-associated proteins compared with those cultured on Matrigel coated onto TCPS (Matrigel_TCPS), resulting in YAP/TEF1 activation and ultimately promoting the transcriptional activities of pancreatic endoderm (PE)-associated genes. Interestingly, YAP activation in PE cells was mediated through integrin α3-FAK-CDC42-PP1A signaling rather than the typical Hippo signaling pathway. Furthermore, pancreatic islet-like organoids (PIOs) generated on Matrigel_PDMS secreted more insulin than those generated from Matrigel_TCPS. Electron micrographs revealed differential Matrigel architectures depending on the underlying substrate, resulting in varying cell-matrix anchorage resistance levels. Accordingly, the high apparent stiffness of the unique mucus-like network structure of Matrigel_PDMS was the critical factor that directly upregulated focal adhesion, thereby leading to better maturation of the pancreatic development of hESCs in vitro.
Collapse
Affiliation(s)
- Eunji Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Tae Yoon Kwon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Youngbin Cho
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Youngjin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yong-Mahn Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
7
|
Hsu MC, Mansouri M, Ahamed NNN, Larson SM, Joshi IM, Ahmed A, Borkholder DA, Abhyankar VV. A miniaturized 3D printed pressure regulator (µPR) for microfluidic cell culture applications. Sci Rep 2022; 12:10769. [PMID: 35750792 PMCID: PMC9232624 DOI: 10.1038/s41598-022-15087-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/17/2022] [Indexed: 01/17/2023] Open
Abstract
Well-defined fluid flows are the hallmark feature of microfluidic culture systems and enable precise control over biophysical and biochemical cues at the cellular scale. Microfluidic flow control is generally achieved using displacement-based (e.g., syringe or peristaltic pumps) or pressure-controlled techniques that provide numerous perfusion options, including constant, ramped, and pulsed flows. However, it can be challenging to integrate these large form-factor devices and accompanying peripherals into incubators or other confined environments. In addition, microfluidic culture studies are primarily carried out under constant perfusion conditions and more complex flow capabilities are often unused. Thus, there is a need for a simplified flow control platform that provides standard perfusion capabilities and can be easily integrated into incubated environments. To this end, we introduce a tunable, 3D printed micro pressure regulator (µPR) and show that it can provide robust flow control capabilities when combined with a battery-powered miniature air pump to support microfluidic applications. We detail the design and fabrication of the µPR and: (i) demonstrate a tunable outlet pressure range relevant for microfluidic applications (1-10 kPa), (ii) highlight dynamic control capabilities in a microfluidic network, (iii) and maintain human umbilical vein endothelial cells (HUVECs) in a multi-compartment culture device under continuous perfusion conditions. We anticipate that our 3D printed fabrication approach and open-access designs will enable customized µPRs that can support a broad range of microfluidic applications.
Collapse
Affiliation(s)
- Meng-Chun Hsu
- Department of Electrical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Nuzhet N N Ahamed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Stephen M Larson
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Indranil M Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - David A Borkholder
- Department of Electrical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA.
| |
Collapse
|
8
|
Dsouza VL, Kuthethur R, Kabekkodu SP, Chakrabarty S. Organ-on-Chip platforms to study tumor evolution and chemosensitivity. Biochim Biophys Acta Rev Cancer 2022; 1877:188717. [PMID: 35304293 DOI: 10.1016/j.bbcan.2022.188717] [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: 10/19/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Despite tremendous advancements in oncology research and therapeutics, cancer remains a primary cause of death worldwide. One of the significant factors in this critical challenge is a precise diagnosis and limited knowledge on how the tumor microenvironment (TME) behaves to the treatment and its role in chemo-resistance. Therefore, it is critical to understand the contribution of a heterogeneous TME in cancer drug response in individual patients for effective therapy management. Micro-physiological systems along with tissue engineering have facilitated the development of more physiologically relevant platforms, known as Organ-on-Chips (OoC). OoC platforms recapitulate the critical hallmarks of the TME in vitro and subsequently abet in sensitivity and efficacy testing of anti-cancer drugs before clinical trials. The OoC platforms incorporating conventional in vitro models enable researchers to control the cellular, molecular, chemical, and biophysical parameters of the TME in precise combinations while analyzing how they contribute to tumor progression and therapy response. This review discusses the application of OoC platforms integrated with conventional 2D cell lines, 3D organoids and spheroid models, and the organotypic tissue slices, including patient-derived and xenograft tumor slice cultures in cancer treatment responses. We summarize the relevance and drawbacks of conventional in vitro models in assessing cancer treatment response, challenges and limitations associated with OoC models, and future opportunities enabled by the OoC technologies towards developing personalized cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Venzil Lavie Dsouza
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raviprasad Kuthethur
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
9
|
Valverde MG, Mille LS, Figler KP, Cervantes E, Li VY, Bonventre JV, Masereeuw R, Zhang YS. Biomimetic models of the glomerulus. Nat Rev Nephrol 2022; 18:241-257. [PMID: 35064233 PMCID: PMC9949601 DOI: 10.1038/s41581-021-00528-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/17/2022]
Abstract
The use of biomimetic models of the glomerulus has the potential to improve our understanding of the pathogenesis of kidney diseases and to enable progress in therapeutics. Current in vitro models comprise organ-on-a-chip, scaffold-based and organoid approaches. Glomerulus-on-a-chip designs mimic components of glomerular microfluidic flow but lack the inherent complexity of the glomerular filtration barrier. Scaffold-based 3D culture systems and organoids provide greater microenvironmental complexity but do not replicate fluid flows and dynamic responses to fluidic stimuli. As the available models do not accurately model the structure or filtration function of the glomerulus, their applications are limited. An optimal approach to glomerular modelling is yet to be developed, but the field will probably benefit from advances in biofabrication techniques. In particular, 3D bioprinting technologies could enable the fabrication of constructs that recapitulate the complex structure of the glomerulus and the glomerular filtration barrier. The next generation of in vitro glomerular models must be suitable for high(er)-content or/and high(er)-throughput screening to enable continuous and systematic monitoring. Moreover, coupling of glomerular or kidney models with those of other organs is a promising approach to enable modelling of partial or full-body responses to drugs and prediction of therapeutic outcomes.
Collapse
Affiliation(s)
- Marta G Valverde
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Luis S Mille
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Kianti P Figler
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Ernesto Cervantes
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Vanessa Y Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Joseph V Bonventre
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA.
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
10
|
Tumour-on-chip microfluidic platform for assessment of drug pharmacokinetics and treatment response. Commun Biol 2021; 4:1001. [PMID: 34429505 PMCID: PMC8385015 DOI: 10.1038/s42003-021-02526-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 08/05/2021] [Indexed: 01/31/2023] Open
Abstract
Microphysiological in vitro systems are platforms for preclinical evaluation of drug effects and significant advances have been made in recent years. However, existing microfluidic devices are not yet able to deliver compounds to cell models in a way that reproduces the real physiological drug exposure. Here, we introduce a novel tumour-on-chip microfluidic system that mimics the pharmacokinetic profile of compounds on 3D tumour spheroids to evaluate their response to the treatments. We used this platform to test the response of SW620 colorectal cancer spheroids to irinotecan (SN38) alone and in combination with the ATM inhibitor AZD0156, using concentrations mimicking mouse plasma exposure profiles of both agents. We explored spheroid volume and viability as a measure of cancer cells response and changes in mechanistically relevant pharmacodynamic biomarkers (γH2AX, cleaved-caspase 3 and Ki67). We demonstrate here that our microfluidic tumour-on-chip platform can successfully predict the efficacy from in vivo studies and therefore represents an innovative tool to guide drug dose and schedules for optimal efficacy and pharmacodynamic assessment, while reducing the need for animal studies. Petreus et al. describe a platform combining 3D tumour-on-chip technology and pump driven microfluidics to study drug effects at physiological exposures preclinically. They test colorectal cancer spheroids with combinations of two drugs and show that it can successfully predict efficacy in vivo, thereby providing a valuable tool for drug response and pharmacodynamic assessment and reducing the need for animal studies.
Collapse
|
11
|
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
|
12
|
Kabandana GKM, Ratajczak AM, Chen C. Making quantitative biomicrofluidics from microbore tubing and 3D-printed adapters. BIOMICROFLUIDICS 2021; 15:034107. [PMID: 34084257 PMCID: PMC8140816 DOI: 10.1063/5.0052314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/12/2021] [Indexed: 06/12/2023]
Abstract
Microfluidic technology has tremendously facilitated the development of in vitro cell cultures and studies. Conventionally, microfluidic devices are fabricated with extensive facilities by well-trained researchers, which hinder the widespread adoption of the technology for broader applications. Enlightened by the fact that low-cost microbore tubing is a natural microfluidic channel, we developed a series of adaptors in a toolkit that can twine, connect, organize, and configure the tubing to produce functional microfluidic units. Three subsets of the toolkit were thoroughly developed: the tubing and scoring tools, the flow adaptors, and the 3D cell culture suite. To demonstrate the usefulness and versatility of the toolkit, we assembled a microfluidic device and successfully applied it for 3D macrophage cultures, flow-based stimulation, and automated near real-time quantitation with new knowledge generated. Overall, we present a new technology that allows simple, fast, and robust assembly of customizable and scalable microfluidic devices with minimal facilities, which is broadly applicable to research that needs or could be enhanced by microfluidics.
Collapse
Affiliation(s)
- Giraso Keza Monia Kabandana
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, USA
| | - Adam Michael Ratajczak
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, USA
| | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, USA
| |
Collapse
|
13
|
Zamprogno P, Thoma G, Cencen V, Ferrari D, Putz B, Michler J, Fantner GE, Guenat OT. Mechanical Properties of Soft Biological Membranes for Organ-on-a-Chip Assessed by Bulge Test and AFM. ACS Biomater Sci Eng 2021; 7:2990-2997. [PMID: 33651947 DOI: 10.1021/acsbiomaterials.0c00515] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Advanced in vitro models called "organ-on-a-chip" can mimic the specific cellular environment found in various tissues. Many of these models include a thin, sometimes flexible, membrane aimed at mimicking the extracellular matrix (ECM) scaffold of in vivo barriers. These membranes are often made of polydimethylsiloxane (PDMS), a silicone rubber that poorly mimics the chemical and physical properties of the basal membrane. However, the ECM and its mechanical properties play a key role in the homeostasis of a tissue. Here, we report about biological membranes with a composition and mechanical properties similar to those found in vivo. Two types of collagen-elastin (CE) membranes were produced: vitrified and nonvitrified (called "hydrogel membrane"). Their mechanical properties were characterized using the bulge test method. The results were compared using atomic force microscopy (AFM), a standard technique used to evaluate the Young's modulus of soft materials at the nanoscale. Our results show that CE membranes with stiffnesses ranging from several hundred of kPa down to 1 kPa can be produced by tuning the CE ratio, the production mode (vitrified or not), and/or certain parameters such as temperature. The Young's modulus can easily be determined using the bulge test. This method is a robust and reproducible to determine membrane stiffness, even for soft membranes, which are more difficult to assess by AFM. Assessment of the impact of substrate stiffness on the spread of human fibroblasts on these surfaces showed that cell spread is lower on softer surfaces than on stiffer surfaces.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Veronika Cencen
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Dario Ferrari
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Barbara Putz
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Johann Michler
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Georg E Fantner
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern 3008, Switzerland.,Department of General Thoracic Surgery, University Hospital of Bern, Bern 3008, Switzerland
| |
Collapse
|
14
|
Patra B, Lateef MA, Brodeur MN, Fleury H, Carmona E, Péant B, Provencher D, Mes-Masson AM, Gervais T. Carboplatin sensitivity in epithelial ovarian cancer cell lines: The impact of model systems. PLoS One 2021; 15:e0244549. [PMID: 33382759 PMCID: PMC7774933 DOI: 10.1371/journal.pone.0244549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/13/2020] [Indexed: 12/26/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy in North America, underscoring the need for the development of new therapeutic strategies for the management of this disease. Although many drugs are pre-clinically tested every year, only a few are selected to be evaluated in clinical trials, and only a small number of these are successfully incorporated into standard care. Inaccuracies with the initial in vitro drug testing may be responsible for some of these failures. Drug testing is often performed using 2D monolayer cultures or 3D spheroid models. Here, we investigate the impact that these different in vitro models have on the carboplatin response of four EOC cell lines, and in particular how different 3D models (polydimethylsiloxane-based microfluidic chips and ultra low attachment plates) influence drug sensitivity within the same cell line. Our results show that carboplatin responses were observed in both the 3D spheroid models tested using apoptosis/cell death markers by flow cytometry. Contrary to previously reported observations, these were not associated with a significant decrease in spheroid size. For the majority of the EOC cell lines (3 out of 4) a similar carboplatin response was observed when comparing both spheroid methods. Interestingly, two cell lines classified as resistant to carboplatin in 2D cultures became sensitive in the 3D models, and one sensitive cell line in 2D culture showed resistance in 3D spheroids. Our results highlight the challenges of choosing the appropriate pre-clinical models for drug testing.
Collapse
Affiliation(s)
- Bishnubrata Patra
- Department of Engineering Physics and Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Muhammad Abdul Lateef
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Melica Nourmoussavi Brodeur
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Hubert Fleury
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Benjamin Péant
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Diane Provencher
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montréal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
- * E-mail: (TG); (AMMM)
| | - Thomas Gervais
- Department of Engineering Physics and Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
- * E-mail: (TG); (AMMM)
| |
Collapse
|
15
|
Three-Dimensional Spheroids as In Vitro Preclinical Models for Cancer Research. Pharmaceutics 2020; 12:pharmaceutics12121186. [PMID: 33291351 PMCID: PMC7762220 DOI: 10.3390/pharmaceutics12121186] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Most cancer biologists still rely on conventional two-dimensional (2D) monolayer culture techniques to test in vitro anti-tumor drugs prior to in vivo testing. However, the vast majority of promising preclinical drugs have no or weak efficacy in real patients with tumors, thereby delaying the discovery of successful therapeutics. This is because 2D culture lacks cell–cell contacts and natural tumor microenvironment, important in tumor signaling and drug response, thereby resulting in a reduced malignant phenotype compared to the real tumor. In this sense, three-dimensional (3D) cultures of cancer cells that better recapitulate in vivo cell environments emerged as scientifically accurate and low cost cancer models for preclinical screening and testing of new drug candidates before moving to expensive and time-consuming animal models. Here, we provide a comprehensive overview of 3D tumor systems and highlight the strategies for spheroid construction and evaluation tools of targeted therapies, focusing on their applicability in cancer research. Examples of the applicability of 3D culture for the evaluation of the therapeutic efficacy of nanomedicines are discussed.
Collapse
|
16
|
Akther F, Yakob SB, Nguyen NT, Ta HT. Surface Modification Techniques for Endothelial Cell Seeding in PDMS Microfluidic Devices. BIOSENSORS 2020; 10:E182. [PMID: 33228050 PMCID: PMC7699314 DOI: 10.3390/bios10110182] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
Microfluidic lab-on-a-chip cell culture techniques have been gaining popularity by offering the possibility of reducing the amount of samples and reagents and greater control over cellular microenvironment. Polydimethylsiloxane (PDMS) is the commonly used polymer for microfluidic cell culture devices because of the cheap and easy fabrication techniques, non-toxicity, biocompatibility, high gas permeability, and optical transparency. However, the intrinsic hydrophobic nature of PDMS makes cell seeding challenging when applied on PDMS surface. The hydrophobicity of the PDMS surface also allows the non-specific absorption/adsorption of small molecules and biomolecules that might affect the cellular behaviour and functions. Hydrophilic modification of PDMS surface is indispensable for successful cell seeding. This review collates different techniques with their advantages and disadvantages that have been used to improve PDMS hydrophilicity to facilitate endothelial cells seeding in PDMS devices.
Collapse
Affiliation(s)
- Fahima Akther
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia;
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
| | - Shazwani Binte Yakob
- School of Pharmacy, the University of Queensland, Brisbane, QLD 4102, Australia;
| | - Nam-Trung Nguyen
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
| | - Hang T. Ta
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia;
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
17
|
Castellanos-Montiel MJ, Velasco I, Escobedo-Avila I. Modeling the neuromuscular junction in vitro: an approach to study neuromuscular junction disorders. Ann N Y Acad Sci 2020; 1488:3-15. [PMID: 33040338 DOI: 10.1111/nyas.14504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized structure that works as an interface to translate the action potential of the presynaptic motor neuron (MN) in the contraction of the postsynaptic myofiber. The design of appropriate experimental models is essential to have efficient and reliable approaches to study NMJ development and function, but also to generate conditions that recapitulate distinct features of diseases. Initial studies relied on the use of tissue slices maintained under the same environment and in which single motor axons were difficult to trace. Later, MNs and muscle cells were obtained from primary cultures or differentiation of progenitors and cocultured as monolayers; however, the tissue architecture was lost. Current approaches include self-assembling 3D structures or the incorporation of biomaterials with cells to generate engineered tissues, although the incorporation of Schwann cells remains a challenge. Thus, numerous investigations have established different NMJ models, some of which are quite complex and challenging. Our review summarizes the in vitro models that have emerged in recent years to coculture MNs and skeletal muscle, trying to mimic the healthy and diseased NMJ. We expect our review may serve as a reference for choosing the appropriate experimental model for the required purposes of investigation.
Collapse
Affiliation(s)
- María José Castellanos-Montiel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.,Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.,Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico City, Mexico
| | - Itzel Escobedo-Avila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
18
|
A Dynamic Hanging-Drop System for Mesenchymal Stem Cell Culture. Int J Mol Sci 2020; 21:ijms21124298. [PMID: 32560269 PMCID: PMC7352343 DOI: 10.3390/ijms21124298] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/02/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
There have been many microfluid technologies combined with hanging-drop for cell culture gotten developed in the past decade. A common problem within these devices is that the cell suspension introduced at the central inlet could cause a number of cells in each microwell to not regularize. Also, the instability of droplets during the spheroid formation remains an unsolved ordeal. In this study, we designed a microfluidic-based hanging-drop culture system with the design of taper-tube that can increase the stability of droplets while enhancing the rate of liquid exchange. A ring is surrounding the taper-tube. The ring can hold the cells to enable us to seed an adequate amount of cells before perfusion. Moreover, during the period of cell culture, the mechanical force around the cell is relatively low to prevent stem cells from differentiate and maintain the phenotype. As a result of our hanging system design, cells are designed to accumulate at the bottom of the droplet. This method enhances convenience for observation activities and analysis of experiments. Thus, this microfluid chip can be used as an in vitro platform representing in vivo physiological conditions, and can be useful in regenerative therapy.
Collapse
|
19
|
Zhu Y, Wu L, Yan H, Lu Z, Yin W, Han H. Enzyme induced molecularly imprinted polymer on SERS substrate for ultrasensitive detection of patulin. Anal Chim Acta 2020; 1101:111-119. [DOI: 10.1016/j.aca.2019.12.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
|
20
|
Affiliation(s)
- Kiran Raj M
- Department of Biomedical EngineeringNational University of Singapore Singapore 117576 Singapore
| | - Suman Chakraborty
- Department of Mechanical EngineeringIndian Institute of Technology Kharagpur Kharagpur 721302 India
| |
Collapse
|
21
|
Williams MJ, Lee NK, Mylott JA, Mazzola N, Ahmed A, Abhyankar VV. A Low-Cost, Rapidly Integrated Debubbler (RID) Module for Microfluidic Cell Culture Applications. MICROMACHINES 2019; 10:mi10060360. [PMID: 31151206 PMCID: PMC6632054 DOI: 10.3390/mi10060360] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 01/08/2023]
Abstract
Microfluidic platforms use controlled fluid flows to provide physiologically relevant biochemical and biophysical cues to cultured cells in a well-defined and reproducible manner. Undisturbed flows are critical in these systems, and air bubbles entering microfluidic channels can lead to device delamination or cell damage. To prevent bubble entry into microfluidic channels, we report a low-cost, Rapidly Integrated Debubbler (RID) module that is simple to fabricate, inexpensive, and easily combined with existing experimental systems. We demonstrate successful removal of air bubbles spanning three orders of magnitude with a maximum removal rate (dV/dt)max = 1.5 mL min−1, at flow rates required to apply physiological wall shear stress (1–200 dyne cm−2) to mammalian cells cultured in microfluidic channels.
Collapse
Affiliation(s)
- Matthew J Williams
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA.
| | - Nicholas K Lee
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA.
| | - Joseph A Mylott
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA.
| | - Nicole Mazzola
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA.
| | - Adeel Ahmed
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA.
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA.
| |
Collapse
|