1
|
She JW, Young CM, Chou SJ, Wu YR, Lin YT, Huang TY, Shen MY, Chen CY, Yang YP, Chien Y, Ayalew H, Liao WH, Tung YC, Shyue JJ, Chiou SH, Yu HH. Gradient conducting polymer surfaces with netrin-1-conjugation promote axon guidance and neuron transmission of human iPSC-derived retinal ganglion cells. Biomaterials 2025; 313:122770. [PMID: 39226653 DOI: 10.1016/j.biomaterials.2024.122770] [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: 05/01/2024] [Revised: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Abstract
Major advances have been made in utilizing human-induced pluripotent stem cells (hiPSCs) for regenerative medicine. Nevertheless, the delivery and integration of hiPSCs into target tissues remain significant challenges, particularly in the context of retinal ganglion cell (RGC) restoration. In this study, we introduce a promising avenue for providing directional guidance to regenerated cells in the retina. First, we developed a technique for construction of gradient interfaces based on functionalized conductive polymers, which could be applied with various functionalized ehthylenedioxythiophene (EDOT) monomers. Using a tree-shaped channel encapsulated with a thin PDMS and a specially designed electrochemical chamber, gradient flow generation could be converted into a functionalized-PEDOT gradient film by cyclic voltammetry. The characteristics of the successfully fabricated gradient flow and surface were analyzed using fluorescent labels, time of flight secondary ion mass spectrometry (TOF-SIMS), and X-ray photoelectron spectroscopy (XPS). Remarkably, hiPSC-RGCs seeded on PEDOT exhibited improvements in neurite outgrowth, axon guidance and neuronal electrophysiology measurements. These results suggest that our novel gradient PEDOT may be used with hiPSC-based technologies as a potential biomedical engineering scaffold for functional restoration of RGCs in retinal degenerative diseases and optic neuropathies.
Collapse
Affiliation(s)
- Jia-Wei She
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan; Taiwan International Graduate Program (TIGP), Nano Science & Technology Program, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan; Department of Engineering and System Science, National Tsing Hua University, No. 101, Section 2, Guangfu Road, East District, 300, Hsinchu City, Taiwan
| | - Chia-Mei Young
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11217, Taiwan
| | - Shih-Jie Chou
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11217, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - You-Ren Wu
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11217, Taiwan
| | - Yu-Ting Lin
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan
| | - Tzu-Yang Huang
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan
| | - Mo-Yuan Shen
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan
| | - Chih-Ying Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Hailemichael Ayalew
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan
| | - Wei-Hao Liao
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Jing-Jong Shyue
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Hwa Chiou
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 11217, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan; Genomic Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| | - Hsiao-Hua Yu
- Smart Organic Materials Laboratory, Institute of Chemistry, Academia Sinica, No. 128, Section 2, Academia Road, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
2
|
Wang D, Feng S, Yang M. Multi-Gradient Bone-Like Nanocomposites Induced by Strain Distribution. ACS NANO 2024; 18:29636-29647. [PMID: 39425938 DOI: 10.1021/acsnano.4c08442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
The heterogeneity of bones is elegantly adapted to the local strain environment, which is critical for maintaining mechanical functions. Such an adaptation enables the strong correlation between strain distributions and multiple gradients, underlying a promising pathway for creating complex gradient structures. However, this potential remains largely unexplored for the synthesis of functional gradient materials. In this work, heterogeneous bone-like nanocomposites with complex structural and compositional gradients comparable to bones are synthesized by inducing strain distributions within the polymer matrix containing amorphous calcium phosphate (ACP). Uniaxial stretching of composite films exerts the highest strain in the center, which ceases gradually toward the sides, resulting in the gradual decrease of polymer alignment and crystallinity. Simultaneously, the center with high orientation traps most ACP during stretching due to the nanoconfinement effect, which in turn promotes the formation of aligned nanofibrous structures. The sides experiencing the least strain have the smallest amounts of ACP, characteristic of porous architectures. Further crystallization of ACP produces oriented apatite nanorods in the center with a larger crystalline/amorphous ratio than the sides because of template-induced crystallization. The combination of structural and compositional gradients leads to gradient mechanical properties, and the gradient span and magnitude correlate nicely with strain distributions. Accompanying bone-like mechanical gradients, the center is less adhesive and self-healable than the sides, which allows a better recovery after a complete cutting. Our work may represent a general strategy for the synthesis of biomimetic materials with complex gradients thanks to the ubiquitous presence of strain distributions in load-bearing structures.
Collapse
Affiliation(s)
- Di Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| | - Ming Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
3
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [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: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
4
|
Suryavanshi P, Bodas D. Knockout cancer by nano-delivered immunotherapy using perfusion-aided scaffold-based tumor-on-a-chip. Nanotheranostics 2024; 8:380-400. [PMID: 38751938 PMCID: PMC11093718 DOI: 10.7150/ntno.87818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/20/2024] [Indexed: 05/18/2024] Open
Abstract
Cancer is a multifactorial disease produced by mutations in the oncogenes and tumor suppressor genes, which result in uncontrolled cell proliferation and resistance to cell death. Cancer progresses due to the escape of altered cells from immune monitoring, which is facilitated by the tumor's mutual interaction with its microenvironment. Understanding the mechanisms involved in immune surveillance evasion and the significance of the tumor microenvironment might thus aid in developing improved therapies. Although in vivo models are commonly utilized, they could be better for time, cost, and ethical concerns. As a result, it is critical to replicate an in vivo model and recreate the cellular and tissue-level functionalities. A 3D cell culture, which gives a 3D architecture similar to that found in vivo, is an appropriate model. Furthermore, numerous cell types can be cocultured, establishing cellular interactions between TME and tumor cells. Moreover, microfluidics perfusion can provide precision flow rates, thus simulating tissue/organ function. Immunotherapy can be used with the perfused 3D cell culture technique to help develop successful therapeutics. Immunotherapy employing nano delivery can target the spot and silence the responsible genes, ensuring treatment effectiveness while minimizing adverse effects. This study focuses on the importance of 3D cell culture in understanding the pathophysiology of 3D tumors and TME, the function of TME in drug resistance, tumor progression, and the development of advanced anticancer therapies for high-throughput drug screening.
Collapse
Affiliation(s)
- Pooja Suryavanshi
- Nanobioscience Group, Agharkar Research Institute, G.G. Agarkar Road, Pune 411 004 India
- Savitribai Phule Pune University, Ganeshkhind Road, Pune 411 007 India
| | - Dhananjay Bodas
- Nanobioscience Group, Agharkar Research Institute, G.G. Agarkar Road, Pune 411 004 India
- Savitribai Phule Pune University, Ganeshkhind Road, Pune 411 007 India
| |
Collapse
|
5
|
Serrano JC, Gillrie MR, Li R, Ishamuddin SH, Moeendarbary E, Kamm RD. Microfluidic-Based Reconstitution of Functional Lymphatic Microvasculature: Elucidating the Role of Lymphatics in Health and Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302903. [PMID: 38059806 PMCID: PMC10837354 DOI: 10.1002/advs.202302903] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/17/2023] [Indexed: 12/08/2023]
Abstract
The knowledge of the blood microvasculature and its functional role in health and disease has grown significantly attributable to decades of research and numerous advances in cell biology and tissue engineering; however, the lymphatics (the secondary vascular system) has not garnered similar attention, in part due to a lack of relevant in vitro models that mimic its pathophysiological functions. Here, a microfluidic-based approach is adopted to achieve precise control over the biological transport of growth factors and interstitial flow that drive the in vivo growth of lymphatic capillaries (lymphangiogenesis). The engineered on-chip lymphatics with in vivo-like morphology exhibit tissue-scale functionality with drainage rates of interstitial proteins and molecules comparable to in vivo standards. Computational and scaling analyses of the underlying transport phenomena elucidate the critical role of the three-dimensional geometry and lymphatic endothelium in recapitulating physiological drainage. Finally, the engineered on-chip lymphatics enabled studies of lymphatic-immune interactions that revealed inflammation-driven responses by the lymphatics to recruit immune cells via chemotactic signals similar to in vivo, pathological events. This on-chip lymphatics platform permits the interrogation of various lymphatic biological functions, as well as screening of lymphatic-based therapies such as interstitial absorption of protein therapeutics and lymphatic immunomodulation for cancer therapy.
Collapse
Affiliation(s)
- Jean C. Serrano
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark R. Gillrie
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medicine University of CalgaryCalgaryABT2N 1N4Canada
| | - Ran Li
- Center for Systems Biology Massachusetts General Hospital Research InstituteBostonMA02114USA
| | - Sarah H. Ishamuddin
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Emad Moeendarbary
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
- 199 Biotechnologies LtdGloucester RoadLondonW2 6LDUK
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
6
|
Lachance GP, Gauvreau D, Boisselier É, Boukadoum M, Miled A. Breaking Barriers: Exploring Neurotransmitters through In Vivo vs. In Vitro Rivalry. SENSORS (BASEL, SWITZERLAND) 2024; 24:647. [PMID: 38276338 PMCID: PMC11154401 DOI: 10.3390/s24020647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
Neurotransmitter analysis plays a pivotal role in diagnosing and managing neurodegenerative diseases, often characterized by disturbances in neurotransmitter systems. However, prevailing methods for quantifying neurotransmitters involve invasive procedures or require bulky imaging equipment, therefore restricting accessibility and posing potential risks to patients. The innovation of compact, in vivo instruments for neurotransmission analysis holds the potential to reshape disease management. This innovation can facilitate non-invasive and uninterrupted monitoring of neurotransmitter levels and their activity. Recent strides in microfabrication have led to the emergence of diminutive instruments that also find applicability in in vitro investigations. By harnessing the synergistic potential of microfluidics, micro-optics, and microelectronics, this nascent realm of research holds substantial promise. This review offers an overarching view of the current neurotransmitter sensing techniques, the advances towards in vitro microsensors tailored for monitoring neurotransmission, and the state-of-the-art fabrication techniques that can be used to fabricate those microsensors.
Collapse
Affiliation(s)
| | - Dominic Gauvreau
- Department Electrical Engineering, Université Laval, Québec, QC G1V 0A6, Canada; (G.P.L.); (D.G.)
| | - Élodie Boisselier
- Department Ophthalmology and Otolaryngology—Head and Neck Surgery, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Mounir Boukadoum
- Department Computer Science, Université du Québec à Montréal, Montréal, QC H2L 2C4, Canada;
| | - Amine Miled
- Department Electrical Engineering, Université Laval, Québec, QC G1V 0A6, Canada; (G.P.L.); (D.G.)
| |
Collapse
|
7
|
Buentello DC, Garcia-Corral M, Trujillo-de Santiago G, Alvarez MM. Neuron(s)-on-a-Chip: A Review of the Design and Use of Microfluidic Systems for Neural Tissue Culture. IEEE Rev Biomed Eng 2024; 17:243-263. [PMID: 36301779 DOI: 10.1109/rbme.2022.3217486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neuron-on-chip (NoC) systems-microfluidic devices in which neurons are cultured-have become a promising alternative to replace or minimize the use of animal models and have greatly facilitated in vitro research. Here, we review and discuss current developments in neuron-on-chip platforms, with a particular emphasis on existing biological models, culturing techniques, biomaterials, and topologies. We also discuss how the architecture, flow, and gradients affect neuronal growth, differentiation, and development. Finally, we discuss some of the most recent applications of NoCs in fundamental research (i.e., studies on the effects of electrical, mechanical/topological, or chemical stimuli) and in disease modeling.
Collapse
|
8
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Liu Y, Guo Q, Zhang X, Wang Y, Mo X, Wu T. Progress in Electrospun Fibers for Manipulating Cell Behaviors. ADVANCED FIBER MATERIALS 2023; 5:1241-1272. [DOI: 10.1007/s42765-023-00281-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/08/2023] [Indexed: 01/06/2025]
|
10
|
Yu J, Yin Y, Leng Y, Zhang J, Wang C, Chen Y, Li X, Wang X, Liu H, Liao Y, Jin Y, Zhang Y, Lu K, Wang K, Wang X, Wang L, Zheng F, Gu Z, Li Y, Fan Y. Emerging strategies of engineering retinal organoids and organoid-on-a-chip in modeling intraocular drug delivery: current progress and future perspectives. Adv Drug Deliv Rev 2023; 197:114842. [PMID: 37105398 DOI: 10.1016/j.addr.2023.114842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Retinal diseases are a rising concern as major causes of blindness in an aging society; therapeutic options are limited, and the precise pathogenesis of these diseases remains largely unknown. Intraocular drug delivery and nanomedicines offering targeted, sustained, and controllable delivery are the most challenging and popular topics in ocular drug development and toxicological evaluation. Retinal organoids (ROs) and organoid-on-a-chip (ROoC) are both emerging as promising in-vitro models to faithfully recapitulate human eyes for retinal research in the replacement of experimental animals and primary cells. In this study, we review the generation and application of ROs resembling the human retina in cell subtypes and laminated structures and introduce the emerging engineered ROoC as a technological opportunity to address critical issues. On-chip vascularization, perfusion, and close inter-tissue interactions recreate physiological environments in vitro, whilst integrating with biosensors facilitates real-time analysis and monitoring during organogenesis of the retina representing engineering efforts in ROoC models. We also emphasize that ROs and ROoCs hold the potential for applications in modeling intraocular drug delivery in vitro and developing next-generation retinal drug delivery strategies.
Collapse
Affiliation(s)
- Jiaheng Yu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yuqi Yin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubing Leng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jingcheng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yanyun Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaorui Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xudong Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yishan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yihan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Keyu Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Kehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Xiaofei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Lizhen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Yubo Fan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| |
Collapse
|
11
|
Blatchley MR, Anseth KS. Middle-out methods for spatiotemporal tissue engineering of organoids. NATURE REVIEWS BIOENGINEERING 2023; 1:329-345. [PMID: 37168734 PMCID: PMC10010248 DOI: 10.1038/s44222-023-00039-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/03/2023] [Indexed: 05/13/2023]
Abstract
Organoids recapitulate many aspects of the complex three-dimensional (3D) organization found within native tissues and even display tissue and organ-level functionality. Traditional approaches to organoid culture have largely employed a top-down tissue engineering strategy, whereby cells are encapsulated in a 3D matrix, such as Matrigel, alongside well-defined biochemical cues that direct morphogenesis. However, the lack of spatiotemporal control over niche properties renders cellular processes largely stochastic. Therefore, bottom-up tissue engineering approaches have evolved to address some of these limitations and focus on strategies to assemble tissue building blocks with defined multi-scale spatial organization. However, bottom-up design reduces the capacity for self-organization that underpins organoid morphogenesis. Here, we introduce an emerging framework, which we term middle-out strategies, that relies on existing design principles and combines top-down design of defined synthetic matrices that support proliferation and self-organization with bottom-up modular engineered intervention to limit the degrees of freedom in the dynamic process of organoid morphogenesis. We posit that this strategy will provide key advances to guide the growth of organoids with precise geometries, structures and function, thereby facilitating an unprecedented level of biomimicry to accelerate the utility of organoids to more translationally relevant applications.
Collapse
Affiliation(s)
- Michael R. Blatchley
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| |
Collapse
|
12
|
Van Breedam E, Ponsaerts P. Promising Strategies for the Development of Advanced In Vitro Models with High Predictive Power in Ischaemic Stroke Research. Int J Mol Sci 2022; 23:ijms23137140. [PMID: 35806146 PMCID: PMC9266337 DOI: 10.3390/ijms23137140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Although stroke is one of the world’s leading causes of death and disability, and more than a thousand candidate neuroprotective drugs have been proposed based on extensive in vitro and animal-based research, an effective neuroprotective/restorative therapy for ischaemic stroke patients is still missing. In particular, the high attrition rate of neuroprotective compounds in clinical studies should make us question the ability of in vitro models currently used for ischaemic stroke research to recapitulate human ischaemic responses with sufficient fidelity. The ischaemic stroke field would greatly benefit from the implementation of more complex in vitro models with improved physiological relevance, next to traditional in vitro and in vivo models in preclinical studies, to more accurately predict clinical outcomes. In this review, we discuss current in vitro models used in ischaemic stroke research and describe the main factors determining the predictive value of in vitro models for modelling human ischaemic stroke. In light of this, human-based 3D models consisting of multiple cell types, either with or without the use of microfluidics technology, may better recapitulate human ischaemic responses and possess the potential to bridge the translational gap between animal-based in vitro and in vivo models, and human patients in clinical trials.
Collapse
|
13
|
Abstract
Embryoids and organoids hold great promise for human biology and medicine. Herein, we discuss conceptual and technological frameworks useful for developing high-fidelity embryoids and organoids that display tissue- and organ-level phenotypes and functions, which are critically needed for decoding developmental programs and improving translational applications. Through dissecting the layers of inputs controlling mammalian embryogenesis, we review recent progress in reconstructing multiscale structural orders in embryoids and organoids. Bioengineering tools useful for multiscale, multimodal structural engineering of tissue- and organ-level cellular organization and microenvironment are also discussed to present integrative, bioengineering-directed approaches to achieve next-generation, high-fidelity embryoids and organoids.
Collapse
Affiliation(s)
- Yue Shao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China; State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
14
|
Synthetic developmental biology: Engineering approaches to guide multicellular organization. Stem Cell Reports 2022; 17:715-733. [PMID: 35276092 PMCID: PMC9023767 DOI: 10.1016/j.stemcr.2022.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022] Open
Abstract
Multicellular organisms of various complexities self-organize in nature. Organoids are in vitro 3D structures that display important aspects of the anatomy and physiology of their in vivo counterparts and that develop from pluripotent or tissue-specific stem cells through a self-organization process. In this review, we describe the multidisciplinary concept of “synthetic developmental biology” where engineering approaches are employed to guide multicellular organization in an experimental setting. We introduce a novel classification of engineering approaches based on the extent of microenvironmental manipulation applied to organoids. In the final section, we discuss how engineering tools might help overcome current limitations in organoid construction.
Collapse
|
15
|
Micro/nanofluidic devices for drug delivery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:9-39. [PMID: 35094782 DOI: 10.1016/bs.pmbts.2021.07.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Micro/nanofluidic drug delivery systems have attracted significant attention as they offer unique advantages in targeted and controlled drug delivery. Based on the desired application, these systems can be categorized into three different groups: in vitro, in situ and in vivo microfluidic drug delivery platforms. In vitro microfluidic drug delivery platforms are closely linked with the emerging concept of lab-on-a-chip for cell culture studies. These systems can be used to administer drugs or therapeutic agents, mostly at the cellular or tissue level, to find the therapeutic index and can potentially be used for personalized medicine. In situ and in vivo microfluidic drug delivery platforms are still at the developmental stage and can be used for drug delivery at tissue or organ levels. A famous example of these systems are microneedles that can be used for painless and controllable delivery of drugs or vaccines through human skin. This chapter presents the cutting edge advances in the design and fabrication of in vitro microfluidic drug delivery systems that can be used for both cellular and tissue drug delivery. It also briefly discusses the in situ drug delivery platforms using microneedles.
Collapse
|
16
|
Roth JG, Huang MS, Li TL, Feig VR, Jiang Y, Cui B, Greely HT, Bao Z, Paşca SP, Heilshorn SC. Advancing models of neural development with biomaterials. Nat Rev Neurosci 2021; 22:593-615. [PMID: 34376834 PMCID: PMC8612873 DOI: 10.1038/s41583-021-00496-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells have emerged as a promising in vitro model system for studying the brain. Two-dimensional and three-dimensional cell culture paradigms have provided valuable insights into the pathogenesis of neuropsychiatric disorders, but they remain limited in their capacity to model certain features of human neural development. Specifically, current models do not efficiently incorporate extracellular matrix-derived biochemical and biophysical cues, facilitate multicellular spatio-temporal patterning, or achieve advanced functional maturation. Engineered biomaterials have the capacity to create increasingly biomimetic neural microenvironments, yet further refinement is needed before these approaches are widely implemented. This Review therefore highlights how continued progression and increased integration of engineered biomaterials may be well poised to address intractable challenges in recapitulating human neural development.
Collapse
Affiliation(s)
- Julien G Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Thomas L Li
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Vivian R Feig
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Yuanwen Jiang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Henry T Greely
- Stanford Law School, Stanford University, Stanford, CA, USA
| | - Zhenan Bao
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
17
|
A microfluidic approach to rescue ALS motor neuron degeneration using rapamycin. Sci Rep 2021; 11:18168. [PMID: 34518579 PMCID: PMC8438029 DOI: 10.1038/s41598-021-97405-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/17/2021] [Indexed: 01/27/2023] Open
Abstract
TAR DNA-binding protein-43 (TDP-43) is known to accumulate in ubiquitinated inclusions of amyotrophic lateral sclerosis affected motor neurons, resulting in motor neuron degeneration, loss of motor functions, and eventually death. Rapamycin, an mTOR inhibitor and a commonly used immunosuppressive drug, has been shown to increase the survivability of Amyotrophic Lateral Sclerosis (ALS) affected motor neurons. Here we present a transgenic, TDP-43-A315T, mouse model expressing an ALS phenotype and demonstrate the presence of ubiquitinated cytoplasmic TDP-43 aggregates with > 80% cell death by 28 days post differentiation in vitro. Embryonic stem cells from this mouse model were used to study the onset, progression, and therapeutic remediation of TDP-43 aggregates using a novel microfluidic rapamycin concentration gradient generator. Results using a microfluidic device show that ALS affected motor neuron survival can be increased by 40.44% in a rapamycin dosage range between 0.4-1.0 µM.
Collapse
|
18
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
19
|
Liu X, Jia Y, Han Z, Hou Q, Zhang W, Zheng W, Jiang X. Integrating a Concentration Gradient Generator and a Single-Cell Trapper Array for High-Throughput Screening the Bioeffects of Nanomaterials. Angew Chem Int Ed Engl 2021; 60:12319-12322. [PMID: 33770418 DOI: 10.1002/anie.202101293] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 02/02/2023]
Abstract
We herein develop a concentration gradient generator (CGG) on a microfluidic chip for diluting different nanoparticles. Specifically designed compact disk (CD)-shaped microchannels in the CGG module could thoroughly mix the flowing solutions and generate a linear concentration gradient of nanoparticles without aggregation. We combine the CGG with a single-cell trapper array (SCA) on microfluidics to evaluate the concentration-dependent bioeffects of the nanoparticles. The precise control of the spatiotemporal generation of nanoparticle concentration on the CGG module and the single-cell-level monitoring of the cell behaviors on the SCA module by a high-content system in real time, render the CGG-SCA system a highly precise platform, which can exclude the average effect of cell population and reflect the response of individual cells to the gradient concentrations accurately. In addition, the CGG-SCA system provides an automated platform for high-throughput screening of nanomedicines with high precision and low sample consumption.
Collapse
Affiliation(s)
- Xiaoyan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, P. R. China.,Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China.,University of Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing, 100049, P. R. China
| | - Yuexiao Jia
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Ziwei Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, P. R. China
| | - Qinghong Hou
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Wei Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing, 100049, P. R. China
| | - Wenfu Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, P. R. China.,University of Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing, 100049, P. R. China
| | - Xingyu Jiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing, 100190, P. R. China.,Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China.,University of Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan District, Beijing, 100049, P. R. China
| |
Collapse
|
20
|
Liu X, Jia Y, Han Z, Hou Q, Zhang W, Zheng W, Jiang X. Integrating a Concentration Gradient Generator and a Single‐Cell Trapper Array for High‐Throughput Screening the Bioeffects of Nanomaterials. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Xiaoyan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for NanoScience and Technology No. 11 Zhongguancun Beiyitiao Beijing 100190 P. R. China
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Road, Nanshan District Shenzhen Guangdong 518055 P. R. China
- Academy for Advanced Interdisciplinary Studies Peking University Beijing 100871 P. R. China
- University of Chinese Academy of Sciences 19 A Yuquan Road, Shijingshan District Beijing 100049 P. R. China
| | - Yuexiao Jia
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Road, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Ziwei Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for NanoScience and Technology No. 11 Zhongguancun Beiyitiao Beijing 100190 P. R. China
| | - Qinghong Hou
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Road, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Wei Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for NanoScience and Technology No. 11 Zhongguancun Beiyitiao Beijing 100190 P. R. China
- University of Chinese Academy of Sciences 19 A Yuquan Road, Shijingshan District Beijing 100049 P. R. China
| | - Wenfu Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for NanoScience and Technology No. 11 Zhongguancun Beiyitiao Beijing 100190 P. R. China
- University of Chinese Academy of Sciences 19 A Yuquan Road, Shijingshan District Beijing 100049 P. R. China
| | - Xingyu Jiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for NanoScience and Technology No. 11 Zhongguancun Beiyitiao Beijing 100190 P. R. China
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Road, Nanshan District Shenzhen Guangdong 518055 P. R. China
- University of Chinese Academy of Sciences 19 A Yuquan Road, Shijingshan District Beijing 100049 P. R. China
| |
Collapse
|
21
|
Spiteri C, Caprettini V, Chiappini C. Biomaterials-based approaches to model embryogenesis. Biomater Sci 2021; 8:6992-7013. [PMID: 33136109 DOI: 10.1039/d0bm01485k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Understanding, reproducing, and regulating the cellular and molecular processes underlying human embryogenesis is critical to improve our ability to recapitulate tissues with proper architecture and function, and to address the dysregulation of embryonic programs that underlies birth defects and cancer. The rapid emergence of stem cell technologies is enabling enormous progress in understanding embryogenesis using simple, powerful, and accessible in vitro models. Biomaterials are playing a central role in providing the spatiotemporal organisation of biophysical and biochemical signalling necessary to mimic, regulate and dissect the evolving embryonic niche in vitro. This contribution is rapidly improving our understanding of the mechanisms underlying embryonic patterning, in turn enabling the development of more effective clinical interventions for regenerative medicine and oncology. Here we highlight how key biomaterial approaches contribute to organise signalling in human embryogenesis models, and we summarise the biological insights gained from these contributions. Importantly, we highlight how nanotechnology approaches have remained largely untapped in this space, and we identify their key potential contributions.
Collapse
Affiliation(s)
- Chantelle Spiteri
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
| | | | | |
Collapse
|
22
|
Samandari M, Rafiee L, Alipanah F, Sanati-Nezhad A, Javanmard SH. A simple, low cost and reusable microfluidic gradient strategy and its application in modeling cancer invasion. Sci Rep 2021; 11:10310. [PMID: 33986379 PMCID: PMC8119451 DOI: 10.1038/s41598-021-89635-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023] Open
Abstract
Microfluidic chemical gradient generators enable precise spatiotemporal control of chemotactic signals to study cellular behavior with high resolution and reliability. However, time and cost consuming preparation steps for cell adhesion in microchannels as well as requirement of pumping facilities usually complicate the application of the microfluidic assays. Here, we introduce a simple strategy for preparation of a reusable and stand-alone microfluidic gradient generator to study cellular behavior. Polydimethylsiloxane (PDMS) is directly mounted on the commercial polystyrene-based cell culture surfaces by manipulating the PDMS curing time to optimize bonding strength. The stand-alone strategy not only offers pumpless application of this microfluidic device but also ensures minimal fluidic pressure and consequently a leakage-free system. Elimination of any surface treatment or coating significantly facilitates the preparation of the microfluidic assay and offers a detachable PDMS microchip which can be reused following to a simple cleaning and sterilization step. The chemotactic signal in our microchip is further characterized using numerical and experimental evaluations and it is demonstrated that the device can generate both linear and polynomial signals. Finally, the feasibility of the strategy in deciphering cellular behavior is demonstrated by exploring cancer cell migration and invasion in response to chemical stimuli. The introduced strategy can significantly decrease the complexity of the microfluidic chemotaxis assays and increase their throughput for various cellular and molecular studies.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Laleh Rafiee
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Fatemeh Alipanah
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Amir Sanati-Nezhad
- Center for Bioengineering Research and Education, and Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Shaghayegh Haghjooy Javanmard
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran.
| |
Collapse
|
23
|
Struzyna LA, Watt ML. The Emerging Role of Neuronal Organoid Models in Drug Discovery: Potential Applications and Hurdles to Implementation. Mol Pharmacol 2021; 99:256-265. [PMID: 33547249 DOI: 10.1124/molpharm.120.000142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
The high failure rate of drugs in the clinical pipeline is likely in part the result of inadequate preclinical models, particularly those for neurologic disorders and neurodegenerative disease. Such preclinical animal models often suffer from fundamental species differences and rarely recapitulate all facets of neurologic conditions, whereas conventional two-dimensional (2D) in vitro models fail to capture the three-dimensional spatial organization and cell-to-cell interactions of brain tissue that are presumed to be critical to the function of the central nervous system. Recent studies have suggested that stem cell-derived neuronal organoids are more physiologically relevant than 2D neuronal cultures because of their cytoarchitecture, electrophysiological properties, human origin, and gene expression. Hence there is interest in incorporating such physiologically relevant models into compound screening and lead optimization efforts within drug discovery. However, despite their perceived relevance, compared with previously used preclinical models, little is known regarding their predictive value. In fact, some have been wary to broadly adopt organoid technology for drug discovery because of the low-throughput and tedious generation protocols, inherent variability, and lack of compatible moderate-to-high-throughput screening assays. Consequently, microfluidic platforms, specialized bioreactors, and automated assays have been and are being developed to address these deficits. This mini review provides an overview of the gaps to broader implementation of neuronal organoids in a drug discovery setting as well as emerging technologies that may better enable their utilization. SIGNIFICANCE STATEMENT: Neuronal organoid models offer the potential for a more physiological system in which to study neurological diseases, and efforts are being made to employ them not only in mechanistic studies but also in profiling/screening purposes within drug discovery. In addition to exploring the utility of neuronal organoid models within this context, efforts in the field aim to standardize such models for consistency and adaptation to screening platforms for throughput evaluation. This review covers potential impact of and hurdles to implementation.
Collapse
|
24
|
Garreta E, Kamm RD, Chuva de Sousa Lopes SM, Lancaster MA, Weiss R, Trepat X, Hyun I, Montserrat N. Rethinking organoid technology through bioengineering. NATURE MATERIALS 2021; 20:145-155. [PMID: 33199860 DOI: 10.1038/s41563-020-00804-4] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 08/18/2020] [Indexed: 06/11/2023]
Abstract
In recent years considerable progress has been made in the development of faithful procedures for the differentiation of human pluripotent stem cells (hPSCs). An important step in this direction has also been the derivation of organoids. This technology generally relies on traditional three-dimensional culture techniques that exploit cell-autonomous self-organization responses of hPSCs with minimal control over the external inputs supplied to the system. The convergence of stem cell biology and bioengineering offers the possibility to provide these stimuli in a controlled fashion, resulting in the development of naturally inspired approaches to overcome major limitations of this nascent technology. Based on the current developments, we emphasize the achievements and ongoing challenges of bringing together hPSC organoid differentiation, bioengineering and ethics. This Review underlines the need for providing engineering solutions to gain control of self-organization and functionality of hPSC-derived organoids. We expect that this knowledge will guide the community to generate higher-grade hPSC-derived organoids for further applications in developmental biology, drug screening, disease modelling and personalized medicine.
Collapse
Affiliation(s)
- Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Roger D Kamm
- Department of Biological Engineering and Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | | | | | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Xavier Trepat
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Insoo Hyun
- Department of Bioethics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for Bioethics, Harvard Medical School, Boston, MA, USA
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
25
|
Aghayee A, Ashton R. Methods for Controlled Induction of Singular Rosette Cytoarchitecture Within Human Pluripotent Stem Cell-Derived Neural Multicellular Assemblies. Methods Mol Biol 2021; 2258:193-203. [PMID: 33340362 DOI: 10.1007/978-1-0716-1174-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Neurally differentiating human pluripotent stem cells (hPSCs) possess the ability to self-organize into structures reminiscent of the developing fetal brain. In 2- and 3D cultures, this phenomenon initiates with formation of polarized areas of neural stem cells (NSCs), known as rosettes that resemble cross-sectional slices of the embryonic neural tube, i.e., the central nervous system (CNS) anlage. Thus, neural rosettes serve as an excellent starting point for bioengineering tissue models of all CNS tissues. Here, we provide detailed methods for bioengineering controlled induction of hPSC-derived neural assemblies with a biomimetic, singular neural rosette cytoarchitecture.
Collapse
Affiliation(s)
- Alireza Aghayee
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Randolph Ashton
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
26
|
Nikolakopoulou P, Rauti R, Voulgaris D, Shlomy I, Maoz BM, Herland A. Recent progress in translational engineered in vitro models of the central nervous system. Brain 2020; 143:3181-3213. [PMID: 33020798 PMCID: PMC7719033 DOI: 10.1093/brain/awaa268] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
The complexity of the human brain poses a substantial challenge for the development of models of the CNS. Current animal models lack many essential human characteristics (in addition to raising operational challenges and ethical concerns), and conventional in vitro models, in turn, are limited in their capacity to provide information regarding many functional and systemic responses. Indeed, these challenges may underlie the notoriously low success rates of CNS drug development efforts. During the past 5 years, there has been a leap in the complexity and functionality of in vitro systems of the CNS, which have the potential to overcome many of the limitations of traditional model systems. The availability of human-derived induced pluripotent stem cell technology has further increased the translational potential of these systems. Yet, the adoption of state-of-the-art in vitro platforms within the CNS research community is limited. This may be attributable to the high costs or the immaturity of the systems. Nevertheless, the costs of fabrication have decreased, and there are tremendous ongoing efforts to improve the quality of cell differentiation. Herein, we aim to raise awareness of the capabilities and accessibility of advanced in vitro CNS technologies. We provide an overview of some of the main recent developments (since 2015) in in vitro CNS models. In particular, we focus on engineered in vitro models based on cell culture systems combined with microfluidic platforms (e.g. 'organ-on-a-chip' systems). We delve into the fundamental principles underlying these systems and review several applications of these platforms for the study of the CNS in health and disease. Our discussion further addresses the challenges that hinder the implementation of advanced in vitro platforms in personalized medicine or in large-scale industrial settings, and outlines the existing differentiation protocols and industrial cell sources. We conclude by providing practical guidelines for laboratories that are considering adopting organ-on-a-chip technologies.
Collapse
Affiliation(s)
- Polyxeni Nikolakopoulou
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Rossana Rauti
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Dimitrios Voulgaris
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Iftach Shlomy
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Anna Herland
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
27
|
Upgrading the Physiological Relevance of Human Brain Organoids. Neuron 2020; 107:1014-1028. [PMID: 32970996 PMCID: PMC10042151 DOI: 10.1016/j.neuron.2020.08.029] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
The recent advent of human pluripotent stem cell (PSC)-derived 3D brain organoids has opened a window into aspects of human brain development that were not accessible before, allowing tractable monitoring and assessment of early developmental processes. However, their broad and effective use for modeling later stages of human brain development and disease is hampered by the lack of a stereotypic anatomical organization, which limits maturation processes dependent upon formation of unique cellular interactions and short- and long-range network connectivity. Emerging methods and technologies aimed at tighter regulatory control through bioengineering approaches, along with newer unbiased organoid analysis readouts, should resolve several of the current limitations. Here, we review recent advances in brain organoid generation and characterization with a focus on highlighting future directions utilizing interdisciplinary strategies that will be important for improving the physiological relevance of this model system.
Collapse
|
28
|
Lowen JM, Leach JK. Functionally graded biomaterials for use as model systems and replacement tissues. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909089. [PMID: 33456431 PMCID: PMC7810245 DOI: 10.1002/adfm.201909089] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 05/03/2023]
Abstract
The heterogeneity of native tissues requires complex materials to provide suitable substitutes for model systems and replacement tissues. Functionally graded materials have the potential to address this challenge by mimicking the gradients in heterogeneous tissues such as porosity, mineralization, and fiber alignment to influence strength, ductility, and cell signaling. Advancements in microfluidics, electrospinning, and 3D printing enable the creation of increasingly complex gradient materials that further our understanding of physiological gradients. The combination of these methods enables rapid prototyping of constructs with high spatial resolution. However, successful translation of these gradients requires both spatial and temporal presentation of cues to model the complexity of native tissues that few materials have demonstrated. This review highlights recent strategies to engineer functionally graded materials for the modeling and repair of heterogeneous tissues, together with a description of how cells interact with various gradients.
Collapse
Affiliation(s)
- Jeremy M. Lowen
- Department of Biomedical Engineering, University of California, Davis, CA, 95616
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, CA, 95616
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817
| |
Collapse
|
29
|
Chen X, Zhang YS, Zhang X, Liu C. Organ-on-a-chip platforms for accelerating the evaluation of nanomedicine. Bioact Mater 2020; 6:1012-1027. [PMID: 33102943 PMCID: PMC7566214 DOI: 10.1016/j.bioactmat.2020.09.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine involves the use of engineered nanoscale materials in an extensive range of diagnostic and therapeutic applications and can be applied to the treatment of many diseases. Despite the rapid progress and tremendous potential of nanomedicine in the past decades, the clinical translational process is still quite slow, owing to the difficulty in understanding, evaluating, and predicting nanomaterial behaviors within the complex environment of human beings. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to resolve these challenges. Sophisticatedly designed Organ Chip enable in vitro simulation of the in vivo microenvironments, thus providing robust platforms for evaluating nanomedicine. Herein, we review recent developments and achievements in Organ Chip models for nanomedicine evaluations, categorized into seven broad sections based on the target organ systems: respiratory, digestive, lymphatic, excretory, nervous, and vascular, as well as coverage on applications relating to cancer. We conclude by providing our perspectives on the challenges and potential future directions for applications of Organ Chip in nanomedicine. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to understand, evaluate, and predict nanomedicine behaviors within the complex environment. Organ Chip models for nanomedicine evaluations are categorized into seven broad sections based on the targeted body systems. Limitations, challenges, and perspectives of Organ Chip for accelerating the assessment of nanomedicine are discussed, respectively.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, United States
| | - Xinping Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
30
|
Chen S, McCarthy A, John JV, Su Y, Xie J. Converting 2D Nanofiber Membranes to 3D Hierarchical Assemblies with Structural and Compositional Gradients Regulates Cell Behavior. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2003754. [PMID: 32944991 PMCID: PMC7606784 DOI: 10.1002/adma.202003754] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/12/2020] [Indexed: 05/24/2023]
Abstract
New methods are described for converting 2D electrospun nanofiber membranes to 3D hierarchical assemblies with structural and compositional gradients. Pore-size gradients are generated by tuning the expansion of 2D membranes in different layers with incorporation of various amounts of a surfactant during the gas-foaming process. The gradient in fiber organizations is formed by expanding 2D nanofiber membranes composed of multiple regions collected by varying rotating speeds of mandrel. A compositional gradient on 3D assemblies consisting of radially aligned nanofibers is prepared by dripping, diffusion, and crosslinking. Bone mesenchymal stem cells (BMSCs) on the 3D nanofiber assemblies with smaller pore size show significantly higher expression of hypoxia-related markers and enhanced chondrogenic differentiation compared to BMSCs cultured on the assemblies with larger pore size. The basic fibroblast growth factor gradient can accelerate fibroblast migration from the surrounding area to the center in an in vitro wound healing model. Taken together, 3D nanofiber assemblies with gradients in pore sizes, fiber organizations, and contents of signaling molecules can be used to engineer tissue constructs for tissue repair and build biomimetic disease models for studying disease biology and screening drugs, in particular, for interface tissue engineering and modeling.
Collapse
Affiliation(s)
- Shixuan Chen
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Alec McCarthy
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Johnson V John
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
31
|
Yamahira S, Satoh T, Yanagawa F, Tamura M, Takagi T, Nakatani E, Kusama Y, Sumaru K, Sugiura S, Kanamori T. Stepwise construction of dynamic microscale concentration gradients around hydrogel-encapsulated cells in a microfluidic perfusion culture device. ROYAL SOCIETY OPEN SCIENCE 2020; 7:200027. [PMID: 32874617 PMCID: PMC7428233 DOI: 10.1098/rsos.200027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/27/2020] [Indexed: 06/11/2023]
Abstract
Inside living organisms, concentration gradients dynamically change over time as biological processes progress. Therefore, methods to construct dynamic microscale concentration gradients in a spatially controlled manner are needed to provide more realistic research environments. Here, we report a novel method for the construction of dynamic microscale concentration gradients in a stepwise manner around cells in micropatterned hydrogel. In our method, cells are encapsulated in a photodegradable hydrogel formed inside a microfluidic perfusion culture device, and perfusion microchannels are then fabricated in the hydrogel by micropatterned photodegradation. The cells in the micropatterned hydrogel can then be cultured by perfusing culture medium through the fabricated microchannels. By using this method, we demonstrate the simultaneous construction of two dynamic concentration gradients, which allowed us to expose the cells encapsulated in the hydrogel to a dynamic microenvironment.
Collapse
Affiliation(s)
- Shinya Yamahira
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Taku Satoh
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Fumiki Yanagawa
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Masato Tamura
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Toshiyuki Takagi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Eri Nakatani
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Yuta Kusama
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Kimio Sumaru
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Shinji Sugiura
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| | - Toshiyuki Kanamori
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba 305-8565, Japan
| |
Collapse
|
32
|
Zhao Y, Demirci U, Chen Y, Chen P. Multiscale brain research on a microfluidic chip. LAB ON A CHIP 2020; 20:1531-1543. [PMID: 32150176 DOI: 10.1039/c9lc01010f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One major challenge in current brain research is generating an integrative understanding of the brain's functions and disorders from its multiscale neuronal architectures and connectivity. Thus, innovative neurotechnology tools are urgently required for deciphering the multiscale functional and structural organizations of the brain at hierarchical scales from the molecular to the organismal level by multiple brain research initiatives launched by the European Union, United States, Australia, Canada, China, Korea, and Japan. To meet this demand, microfluidic chips (μFCs) have rapidly evolved as a trans-scale neurotechnological toolset to enable multiscale studies of the brain due to their unique advantages in flexible microstructure design, multifunctional integration, accurate microenvironment control, and capacity for automatic sample processing. Here, we review the recent progress in applying innovative μFC-based neuro-technologies to promote multiscale brain research and uniquely focus on representative applications of μFCs to address challenges in brain research at each hierarchical level. We discuss the current trend of combinational applications of μFCs with other neuro- and biotechnologies, including optogenetics, brain organoids, and 3D bioprinting, for better multiscale brain research. In addition, we offer our insights into the existing outstanding questions at each hierarchical level of brain research that could potentially be addressed by advancing microfluidic techniques. This review will serve as a timely guide for bioengineers and neuroscientists to develop and apply μFC-based neuro-technologies for promoting basic and translational brain research.
Collapse
Affiliation(s)
- Yanan Zhao
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, 115 Donghu Road, Wuhan 430071, China.
| | | | | | | |
Collapse
|
33
|
|
34
|
Microfluidic Systems in CNS Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32285366 DOI: 10.1007/978-3-030-36588-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Current technological progress facilitates the introduction of micro-devices into biotechnology research including studies on central nervous system. Wide range of micro-chambers with diversity of channel systems and multiple compartments enable users to create models which closely mimic nervous tissue structure which nowadays is often called as brain-on-a-chip technology. Heretofore experiments showing the influence of substance gradients, cell interactions, spatial conditions, neuroinflammation, stem cells migration, drug delivery, mechanisms controlling progression of diseases like Alzheimer, Parkinson, multiple sclerosis or nerve injury were performed in microfluidic devices. Moreover, the integration of bio-sensors and development of dedicated software for microfluidic studies can enable performing high throughput and good quality automated experiments investigating regeneration and degeneration processes in models well emulating central nervous system structures.
Collapse
|
35
|
Chemotactic Responses of Jurkat Cells in Microfluidic Flow-Free Gradient Chambers. MICROMACHINES 2020; 11:mi11040384. [PMID: 32260431 PMCID: PMC7231302 DOI: 10.3390/mi11040384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/29/2022]
Abstract
Gradients of soluble molecules coordinate cellular communication in a diverse range of multicellular systems. Chemokine-driven chemotaxis is a key orchestrator of cell movement during organ development, immune response and cancer progression. Chemotaxis assays capable of examining cell responses to different chemokines in the context of various extracellular matrices will be crucial to characterize directed cell motion in conditions which mimic whole tissue conditions. Here, a microfluidic device which can generate different chemokine patterns in flow-free gradient chambers while controlling surface extracellular matrix (ECM) to study chemotaxis either at the population level or at the single cell level with high resolution imaging is presented. The device is produced by combining additive manufacturing (AM) and soft lithography. Generation of concentration gradients in the device were simulated and experimentally validated. Then, stable gradients were applied to modulate chemotaxis and chemokinetic response of Jurkat cells as a model for T lymphocyte motility. Live imaging of the gradient chambers allowed to track and quantify Jurkat cell migration patterns. Using this system, it has been found that the strength of the chemotactic response of Jurkat cells to CXCL12 gradient was reduced by increasing surface fibronectin in a dose-dependent manner. The chemotaxis of the Jurkat cells was also found to be governed not only by the CXCL12 gradient but also by the average CXCL12 concentration. Distinct migratory behaviors in response to chemokine gradients in different contexts may be physiologically relevant for shaping the host immune response and may serve to optimize the targeting and accumulation of immune cells to the inflammation site. Our approach demonstrates the feasibility of using a flow-free gradient chamber for evaluating cross-regulation of cell motility by multiple factors in different biologic processes.
Collapse
|
36
|
O'Grady BJ, Lippmann ES. Recent Advancements in Engineering Strategies for Manipulating Neural Stem Cell Behavior. ACTA ACUST UNITED AC 2020; 1:41-47. [PMID: 33748772 DOI: 10.1007/s43152-020-00003-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Purpose of Review Stem cells are exquisitely sensitive to biophysical and biochemical cues within the native microenvironment. This review focuses on emerging strategies to manipulate neural cell behavior using these influences in three-dimensional (3D) culture systems. Recent Findings Traditional systems for neural cell differentiation typically produce heterogeneous populations with limited diversity rather than the complex, organized tissue structures observed in vivo. Advancements in developing engineering tools to direct neural cell fates can enable new applications in basic research, disease modeling, and regenerative medicine. Summary This review article highlights engineering strategies that facilitate controlled presentation of biophysical and biochemical cues to guide differentiation and impart desired phenotypes on neural cell populations. Specific highlighted examples include engineered biomaterials and microfluidic platforms for spatiotemporal control over the presentation of morphogen gradients.
Collapse
Affiliation(s)
- Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
37
|
Koens R, Tabata Y, Serrano JC, Aratake S, Yoshino D, Kamm RD, Funamoto K. Microfluidic platform for three-dimensional cell culture under spatiotemporal heterogeneity of oxygen tension. APL Bioeng 2020; 4:016106. [PMID: 32161836 PMCID: PMC7060087 DOI: 10.1063/1.5127069] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Cells in a tumor microenvironment are exposed to spatial and temporal variations in oxygen tension due to hyperproliferation and immature vascularization. Such spatiotemporal oxygen heterogeneity affects the behavior of cancer cells, leading to cancer growth and metastasis, and thus, it is essential to clarify the cellular responses of cancer cells to oxygen tension. Herein, we describe a new double-layer microfluidic device allowing the control of oxygen tension and the behavior of cancer cells under spatiotemporal oxygen heterogeneity. Two parallel gas channels were located above the media and gel channels to enhance gas exchange, and a gas-impermeable polycarbonate film was embedded in the device to prevent the diffusion of atmospheric oxygen. Variations in oxygen tension in the device with the experimental parameters and design variables were investigated computationally and validated by using oxygen-sensitive nanoparticles. The present device can generate a uniform hypoxic condition at oxygen levels down to 0.3% O2, as well as a linear oxygen gradient from 3% O2 to 17% O2 across the gel channel within 15 min. Moreover, human breast cancer cells suspended in type I collagen gel were introduced in the gel channel to observe their response under controlled oxygen tension. Hypoxic exposure activated the proliferation and motility of the cells, which showed a local maximum increase at 5% O2. Under the oxygen gradient condition, the increase in the cell number was relatively high in the central mild hypoxia region. These findings demonstrate the utility of the present device to study cellular responses in an oxygen-controlled microenvironment.
Collapse
Affiliation(s)
- Rei Koens
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-12 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | | | - Jean C. Serrano
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA
| | | | | | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
38
|
Shen S, Zhang X, Zhang F, Wang D, Long D, Niu Y. Three-gradient constructions in a flow-rate insensitive microfluidic system for drug screening towards personalized treatment. Talanta 2020; 208:120477. [DOI: 10.1016/j.talanta.2019.120477] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022]
|
39
|
Mo SJ, Lee JH, Kye HG, Lee JM, Kim EJ, Geum D, Sun W, Chung BG. A microfluidic gradient device for drug screening with human iPSC-derived motoneurons. Analyst 2020; 145:3081-3089. [DOI: 10.1039/c9an02384d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We developed a microfluidic gradient device to utilize as a drug screening system with human induced pluripotent stem cell (hiPSC)-derived motoneurons.
Collapse
Affiliation(s)
- Sung Joon Mo
- Department of Biomedical Engineering
- Sogang University
- Korea
| | - Ju-Hyun Lee
- Department of Anatomy
- Brain Korea 21 Program
- Korea University College of Medicine
- Seoul
- Korea
| | - Hyeon Gi Kye
- Department of Mechanical Engineering
- Sogang University
- Seoul
- Korea
| | - Jong Min Lee
- Department of Mechanical Engineering
- Sogang University
- Seoul
- Korea
| | | | - Dongho Geum
- Department of Biomedical Sciences
- Korea University College of Medicine
- Seoul
- Korea
| | - Woong Sun
- Department of Anatomy
- Brain Korea 21 Program
- Korea University College of Medicine
- Seoul
- Korea
| | - Bong Geun Chung
- Department of Mechanical Engineering
- Sogang University
- Seoul
- Korea
| |
Collapse
|
40
|
Tunesi M, Izzo L, Raimondi I, Albani D, Giordano C. A miniaturized hydrogel-based in vitro model for dynamic culturing of human cells overexpressing beta-amyloid precursor protein. J Tissue Eng 2020; 11:2041731420945633. [PMID: 32922719 PMCID: PMC7446262 DOI: 10.1177/2041731420945633] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
Recent findings have highlighted an interconnection between intestinal microbiota and the brain, referred to as microbiota-gut-brain axis, and suggested that alterations in microbiota composition might affect brain functioning, also in Alzheimer's disease. To investigate microbiota-gut-brain axis biochemical pathways, in this work we developed an innovative device to be used as modular unit in an engineered multi-organ-on-a-chip platform recapitulating in vitro the main players of the microbiota-gut-brain axis, and an innovative three-dimensional model of brain cells based on collagen/hyaluronic acid or collagen/poly(ethylene glycol) semi-interpenetrating polymer networks and β-amyloid precursor protein-Swedish mutant-expressing H4 cells, to simulate the pathological scenario of Alzheimer's disease. We set up the culturing conditions, assessed cell response, scaled down the three-dimensional models to be hosted in the organ-on-a-chip device, and cultured them both in static and in dynamic conditions. The results suggest that the device and three-dimensional models are exploitable for advanced engineered models representing brain features also in Alzheimer's disease scenario.
Collapse
Affiliation(s)
- Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
| | - Ilaria Raimondi
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri – IRCSS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
| |
Collapse
|
41
|
|
42
|
Pena CD, Zhang S, Majeska R, Venkatesh T, Vazquez M. Invertebrate Retinal Progenitors as Regenerative Models in a Microfluidic System. Cells 2019; 8:cells8101301. [PMID: 31652654 PMCID: PMC6829900 DOI: 10.3390/cells8101301] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/30/2022] Open
Abstract
Regenerative retinal therapies have introduced progenitor cells to replace dysfunctional or injured neurons and regain visual function. While contemporary cell replacement therapies have delivered retinal progenitor cells (RPCs) within customized biomaterials to promote viability and enable transplantation, outcomes have been severely limited by the misdirected and/or insufficient migration of transplanted cells. RPCs must achieve appropriate spatial and functional positioning in host retina, collectively, to restore vision, whereas movement of clustered cells differs substantially from the single cell migration studied in classical chemotaxis models. Defining how RPCs interact with each other, neighboring cell types and surrounding extracellular matrixes are critical to our understanding of retinogenesis and the development of effective, cell-based approaches to retinal replacement. The current article describes a new bio-engineering approach to investigate the migratory responses of innate collections of RPCs upon extracellular substrates by combining microfluidics with the well-established invertebrate model of Drosophila melanogaster. Experiments utilized microfluidics to investigate how the composition, size, and adhesion of RPC clusters on defined extracellular substrates affected migration to exogenous chemotactic signaling. Results demonstrated that retinal cluster size and composition influenced RPC clustering upon extracellular substrates of concanavalin (Con-A), Laminin (LM), and poly-L-lysine (PLL), and that RPC cluster size greatly altered collective migratory responses to signaling from Fibroblast Growth Factor (FGF), a primary chemotactic agent in Drosophila. These results highlight the significance of examining collective cell-biomaterial interactions on bio-substrates of emerging biomaterials to aid directional migration of transplanted cells. Our approach further introduces the benefits of pairing genetically controlled models with experimentally controlled microenvironments to advance cell replacement therapies.
Collapse
Affiliation(s)
- Caroline D Pena
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA.
| | - Stephanie Zhang
- Department of Biomedical Engineering, The State University of New York at Binghamton, NY 13902, USA.
| | - Robert Majeska
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA.
| | - Tadmiri Venkatesh
- Department of Biology, City College of New York, New York, NY 10031, USA.
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers University, The State University of New Jersey, New Brunswick, NJ 08854, USA.
| |
Collapse
|
43
|
Engineered signaling centers for the spatially controlled patterning of human pluripotent stem cells. Nat Methods 2019; 16:640-648. [DOI: 10.1038/s41592-019-0455-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 05/15/2019] [Indexed: 12/29/2022]
|
44
|
Regier MC, Tokar JJ, Warrick JW, Pabon L, Berthier E, Beebe DJ, Stevens KR. User-defined morphogen patterning for directing human cell fate stratification. Sci Rep 2019; 9:6433. [PMID: 31015521 PMCID: PMC6478938 DOI: 10.1038/s41598-019-42874-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Concentration gradients of biochemical stimuli such as morphogens play a critical role in directing cell fate patterning across species and throughout development but are not commonly recapitulated in vitro. While in vitro biomolecule gradients have been generated using customized microfluidic platforms, broad implementation has been limited because these platforms introduce new variables to cell culture such as externally driven flow, culture in a specialized matrix, or extended time for in situ long range diffusion. Here we introduce a method that enables preforming and then transferring user-controlled gradients to cells in standard "open" cultures. Our gradient patterning devices are modular and decoupled from the culture substrate. We find that gradient generation and transfer are predictable by finite element modeling and that device and loading parameters can be used to tune the stimulus pattern. Furthermore, we demonstrate use of these devices to spatially define morphogen signal gradients and direct peri-gastrulation fate stratification of human pluripotent stem cells. This method for extrinsic application of biochemical signal gradients can thus be used to spatially influence cellular fate decisions in a user-controlled manner.
Collapse
Affiliation(s)
- Mary C Regier
- Department of Bioengineering, University of Washington, 98195, Seattle, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jacob J Tokar
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin - Madison, 53705, Madison, USA
| | - Lil Pabon
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Pathology, University of Washington, 98195, Seattle, USA
| | - Erwin Berthier
- Department of Chemistry, University of Washington, 98195, Seattle, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, 98195, Seattle, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA.
- Department of Pathology, University of Washington, 98195, Seattle, USA.
| |
Collapse
|
45
|
Wulftange WJ, Rose MA, Garmendia-Cedillos M, da Silva D, Poprawski JE, Srinivasachar D, Sullivan T, Lim L, Bliskovsky VV, Hall MD, Pohida TJ, Robey RW, Morgan NY, Gottesman MM. Spatial control of oxygen delivery to three-dimensional cultures alters cancer cell growth and gene expression. J Cell Physiol 2019; 234:20608-20622. [PMID: 31012116 DOI: 10.1002/jcp.28665] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/22/2019] [Indexed: 01/01/2023]
Abstract
Commonly used monolayer cancer cell cultures fail to provide a physiologically relevant environment in terms of oxygen delivery. Here, we describe a three-dimensional (3D) bioreactor system where cancer cells are grown in Matrigel in modified six-well plates. Oxygen is delivered to the cultures through a polydimethylsiloxane (PDMS) membrane at the bottom of the wells, with microfabricated PDMS pillars to control oxygen delivery. The plates receive 3% oxygen from below and 0% oxygen at the top surface of the media, providing a gradient of 3-0% oxygen. We compared growth and transcriptional profiles for cancer cells grown in Matrigel in the bioreactor, 3D cultures grown in 21% oxygen, and cells grown in a standard hypoxia chamber at 3% oxygen. Additionally, we compared gene expression of conventional two-dimensional monolayer culture and 3D Matrigel culture in 21% oxygen. We conclude that controlled oxygen delivery may provide a more physiologically relevant 3D system.
Collapse
Affiliation(s)
- William J Wulftange
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland.,Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle A Rose
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Marcial Garmendia-Cedillos
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, Maryland
| | - Davi da Silva
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Joanna E Poprawski
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dhruv Srinivasachar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Taylor Sullivan
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Langston Lim
- Confocal Microscopy Core Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Valery V Bliskovsky
- CCR Genomics Core, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Thomas J Pohida
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nicole Y Morgan
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
46
|
Silva TP, Cotovio JP, Bekman E, Carmo-Fonseca M, Cabral JMS, Fernandes TG. Design Principles for Pluripotent Stem Cell-Derived Organoid Engineering. Stem Cells Int 2019; 2019:4508470. [PMID: 31149014 PMCID: PMC6501244 DOI: 10.1155/2019/4508470] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 02/24/2019] [Indexed: 12/17/2022] Open
Abstract
Human morphogenesis is a complex process involving distinct microenvironmental and physical signals that are manipulated in space and time to give rise to complex tissues and organs. Advances in pluripotent stem cell (PSC) technology have promoted the in vitro recreation of processes involved in human morphogenesis. The development of organoids from human PSCs represents one reliable source for modeling a large spectrum of human disorders, as well as a promising approach for drug screening and toxicological tests. Based on the "self-organization" capacity of stem cells, different PSC-derived organoids have been created; however, considerable differences between in vitro-generated PSC-derived organoids and their in vivo counterparts have been reported. Advances in the bioengineering field have allowed the manipulation of different components, including cellular and noncellular factors, to better mimic the in vivo microenvironment. In this review, we focus on different examples of bioengineering approaches used to promote the self-organization of stem cells, including assembly, patterning, and morphogenesis in vitro, contributing to tissue-like structure formation.
Collapse
Affiliation(s)
- Teresa P. Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - João P. Cotovio
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia Bekman
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - Maria Carmo-Fonseca
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
47
|
Mobini S, Song YH, McCrary MW, Schmidt CE. Advances in ex vivo models and lab-on-a-chip devices for neural tissue engineering. Biomaterials 2019; 198:146-166. [PMID: 29880219 PMCID: PMC6957334 DOI: 10.1016/j.biomaterials.2018.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/25/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023]
Abstract
The technologies related to ex vivo models and lab-on-a-chip devices for studying the regeneration of brain, spinal cord, and peripheral nerve tissues are essential tools for neural tissue engineering and regenerative medicine research. The need for ex vivo systems, lab-on-a-chip technologies and disease models for neural tissue engineering applications are emerging to overcome the shortages and drawbacks of traditional in vitro systems and animal models. Ex vivo models have evolved from traditional 2D cell culture models to 3D tissue-engineered scaffold systems, bioreactors, and recently organoid test beds. In addition to ex vivo model systems, we discuss lab-on-a-chip devices and technologies specifically for neural tissue engineering applications. Finally, we review current commercial products that mimic diseased and normal neural tissues, and discuss the future directions in this field.
Collapse
Affiliation(s)
- Sahba Mobini
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Michaela W McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
48
|
O'Grady B, Balikov DA, Wang JX, Neal EK, Ou YC, Bardhan R, Lippmann ES, Bellan LM. Spatiotemporal control and modeling of morphogen delivery to induce gradient patterning of stem cell differentiation using fluidic channels. Biomater Sci 2019; 7:1358-1371. [PMID: 30778445 PMCID: PMC6485939 DOI: 10.1039/c8bm01199k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The process of cell differentiation in a developing embryo is influenced by numerous factors, including various biological molecules whose presentation varies dramatically over space and time. These morphogens regulate cell fate based on concentration profiles, thus creating discrete populations of cells and ultimately generating large, complex tissues and organs. Recently, several in vitro platforms have attempted to recapitulate the complex presentation of extrinsic signals found in nature. However, it has been a challenge to design versatile platforms that can dynamically control morphogen gradients over extended periods of time. To address some of these issues, we introduce a platform using channels patterned in hydrogels to deliver multiple morphogens to cells in a 3D scaffold, thus creating a spectrum of cell phenotypes based on the resultant morphogen gradients. The diffusion coefficient of a common small molecule morphogen, retinoic acid (RA), was measured within our hydrogel platform using Raman spectroscopy and its diffusion in our platform's geometry was modeled using finite element analysis. The predictive model of spatial gradients was validated in a cell-free hydrogel, and temporal control of morphogen gradients was then demonstrated using a reporter cell line that expresses green fluorescent protein in the presence of RA. Finally, the utility of this approach for regulating cell phenotype was demonstrated by generating opposing morphogen gradients to create a spectrum of mesenchymal stem cell differentiation states.
Collapse
Affiliation(s)
- Brian O'Grady
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
George J, Hsu CC, Nguyen LTB, Ye H, Cui Z. Neural tissue engineering with structured hydrogels in CNS models and therapies. Biotechnol Adv 2019; 42:107370. [PMID: 30902729 DOI: 10.1016/j.biotechadv.2019.03.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/25/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
Abstract
The development of techniques to create and use multiphase microstructured hydrogels (granular hydrogels or microgels) has enabled the generation of cultures with more biologically relevant architecture and use of structured hydrogels is especially pertinent to the development of new types of central nervous system (CNS) culture models and therapies. We review material choice and the customisation of hydrogel structure, as well as the use of hydrogels in developmental models. Combining the use of structured hydrogel techniques with developmentally relevant tissue culture approaches will enable the generation of more relevant models and treatments to repair damaged CNS tissue architecture.
Collapse
Affiliation(s)
- Julian George
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Chia-Chen Hsu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Linh Thuy Ba Nguyen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
50
|
Kim S, Masum F, Kim JK, Chung HJ, Jeon JS. On-chip phenotypic investigation of combinatory antibiotic effects by generating orthogonal concentration gradients. LAB ON A CHIP 2019; 19:959-973. [PMID: 30768106 DOI: 10.1039/c8lc01406j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Combinatory therapy using two or more kinds of antibiotics is attracting considerable attention for inhibiting multi-drug resistant pathogenic bacteria. Although the therapy mostly leads to more powerful antimicrobial effects than using a single antibiotic (synergy), interference may arise from certain antibiotic combinations, resulting in the antimicrobial effect being suppressed (antagonism). Here, we present a microfluidic-based phenotypic screening chip to investigate combinatory antibiotic effects by automatically generating two orthogonal concentration gradients on a bacteria-trapping agarose gel. Computational simulations and fluorescence experiments together verify the simultaneous establishment of 121 concentration combinations, facilitating on-chip drug testing with stability and efficiency. Against Gram-negative bacteria, Pseudomonas aeruginosa, our chip allows the measurement of phenotypic growth levels, and enables various types of analyses for all antibiotic pairs to be conducted in 7 h. Furthermore, by providing a specific amount of susceptibility data, our chip enables the two reference models, Loewe additivity and Bliss independence, to be implemented, which classify the antibiotic interaction types into synergy or antagonism. These results suggest the efficacy of our chip as a cell-based drug screening platform for exploring the underlying pharmacological patterns of antibiotic interactions, with potential applications in guidance in clinical therapies and in screening other cell-type agents.
Collapse
Affiliation(s)
- Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| | | | | | | | | |
Collapse
|