1
|
Zhang J, Fu Y, Fong CY, Hua H, Li W, Khoo BL. Advancements in microfluidic technology for rapid bacterial detection and inflammation-driven diseases. LAB ON A CHIP 2025. [PMID: 40201957 DOI: 10.1039/d4lc00795f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Bacterial detection is pivotal for the timely diagnosis and effective treatment of infectious diseases. Microfluidic platforms offer advantages over traditional methods, including heightened sensitivity, rapid analysis, and minimal sample volume requirements. Traditional clinical methods for bacterial identification often involve extended processing times and necessitate high pathogen concentrations, resulting in delayed diagnoses and missed treatment opportunities. Microfluidic technology overcomes these limitations by facilitating rapid bacterial identification at lower biomass levels, thus ensuring prompt and precise treatment interventions. Additionally, bacteria-driven inflammation has been associated with the development and progression of various diseases, including cancer. Elucidating the complex interplay between bacteria, inflammation, and disease is essential for devising effective disease models and therapeutic strategies. Microfluidic platforms have been used to construct in vitro disease models that accurately replicate the intricate microenvironment that bacteria-driven inflammation affects. These models offer valuable insights into bacteria-driven inflammation and its impact on disease progression, such as cancer metastasis and therapeutic responses. This review examines recent advancements in bacterial detection using microfluidics and assesses the potential of this technology as a robust tool for exploring bacteria-driven inflammation in the context of cancer.
Collapse
Affiliation(s)
- Jing Zhang
- College of Basic Medicine, Hebei University, Baoding, China
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-autoimmune Diseases in Hebei Province, Hebei University, Baoding 071000, China
| | - Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Ching Yin Fong
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
| | - Haojun Hua
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
| | - Wei Li
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen-Futian Research Institute, Shenzhen 518057, China
| |
Collapse
|
2
|
Yoshida A, Baba K, Takahashi H, Nagese K, Shimizu T. One-step fabrication of 3D-aligned human skeletal muscle tissue and measurement of contractile force for preclinical drug testing. Mater Today Bio 2025; 31:101456. [PMID: 39896285 PMCID: PMC11783003 DOI: 10.1016/j.mtbio.2025.101456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Human muscle tissue models are critical to understanding the complex physiology of skeletal muscle in studies of drug discovery, development, and toxicity profiling in the human body. However, due to the challenges in in vitro maturation of human muscle cells, few research groups developing their own tissue engineering techniques have succeeded in producing contractile human muscle tissues. Moreover, a more sophisticated method is necessary to measure contractile forces generated by the muscle tissues for preclinical studies in muscle physiology and drug discovery. Although a few research groups have established their own tissue model systems that measure contractile force, they require multi-step fabrication processes to produce human muscle tissues sufficiently functional to be able to measure the contractile forces. To improve the usability of our tissue model system, this study focused on simplifying the tissue engineering approach to produce a practical muscle tissue model. In this study, muscle satellite cells were simply mixed with a combination of fibrinogen, thrombin, and Matrigel before gel formation. The presence of muscle satellite cells induces gel compaction and spontaneously induces unidirectional stretching of the gel, resulting in the muscle satellite cells being aligned three-dimensionally with the direction of stretching. Furthermore, this gel environment promotes the maturation of the human muscle progenitor cells into aligned myofibers, also provides the tissue with an elastic platform for muscle contraction, and allows the attachment of the muscle tissue to a device for measurement of contractile force. Therefore, this one-step tissue fabrication allowed us to produce 3D-aligned human muscle tissues and this tissue model is ready to use for the measurement of contractile forces. In fact, the muscle contractions created by electrical and chemical stimulation were quantitatively determined using our measurement system. In addition, the impact of some representative drugs on this muscle tissue were able to be monitored in real-time throughout the changes in contractile forces. In conclusion, our tissue model system, produced by a simple fabrication method, can be used for preclinical in vitro studies in muscle physiology and drug discovery.
Collapse
Affiliation(s)
- Azumi Yoshida
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Kazuki Baba
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Kenichi Nagese
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
- Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| |
Collapse
|
3
|
Shi K, He C, Pan H, Liu D, Zhang J, Han W, Xiang Y, Hu N. Advanced passive 3D bioelectronics: powerful tool for the cardiac electrophysiology investigation. MICROSYSTEMS & NANOENGINEERING 2025; 11:50. [PMID: 40097396 PMCID: PMC11914486 DOI: 10.1038/s41378-025-00891-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/21/2025] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Cardiovascular diseases (CVDs) are the first cause of death globally, posing a significant threat to human health. Cardiac electrophysiology is pivotal for the understanding and management of CVDs, particularly for addressing arrhythmias. A significant proliferation of micro-nano bioelectric devices and systems has occurred in the field of cardiomyocyte electrophysiology. These bioelectronic platforms feature distinctive electrode geometries that improve the fidelity of native electrophysiological signals. Despite the prevalence of planar microelectrode arrays (MEAs) for simultaneous multichannel recording of cellular electrophysiological signals, extracellular recordings often yield suboptimal signal quality. In contrast, three-dimensional (3D) MEAs and advanced penetration strategies allow high-fidelity intracellular signal detection. 3D nanodevices are categorized into the active and the passive. Active devices rely on external power sources to work, while passive devices operate without external power. Passive devices possess simplicity, biocompatibility, stability, and lower power consumption compared to active ones, making them ideal for sensors and implantable applications. This review comprehensively discusses the fabrication, geometric configuration, and penetration strategies of passive 3D micro/nanodevices, emphasizing their application in drug screening and disease modeling. Moreover, we summarize existing challenges and future opportunities to develop passive micro/nanobioelectronic devices from cardiac electrophysiological research to cardiovascular clinical practice.
Collapse
Affiliation(s)
- Keda Shi
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chengwen He
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China
| | - Hui Pan
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Dong Liu
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ji Zhang
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Weili Han
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Yuting Xiang
- Department of Obstetrics, the Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, China.
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China.
| |
Collapse
|
4
|
Verdugo-Avello F, Wychowaniec JK, Villacis-Aguirre CA, D'Este M, Toledo JR. Bone microphysiological models for biomedical research. LAB ON A CHIP 2025; 25:806-836. [PMID: 39906932 DOI: 10.1039/d4lc00762j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Bone related disorders are highly prevalent, and many of these pathologies still do not have curative and definitive treatment methods. This is due to a complex interplay of multiple factors, such as the crosstalk between different tissues and cellular components, all of which are affected by microenvironmental factors. Moreover, these bone pathologies are specific, and current treatment results vary from patient to patient owing to their intrinsic biological variability. Current approaches in drug development to deliver new drug candidates against common bone disorders, such as standard two-dimensional (2D) cell culture and animal-based studies, are now being replaced by more relevant diseases modelling, such as three-dimension (3D) cell culture and primary cells under human-focused microphysiological systems (MPS) that can resemble human physiology by mimicking 3D tissue organization and cell microenvironmental cues. In this review, various technological advancements for in vitro bone modeling are discussed, highlighting the progress in biomaterials used as extracellular matrices, stem cell biology, and primary cell culture techniques. With emphasis on examples of modeling healthy and disease-associated bone tissues, this tutorial review aims to survey current approaches of up-to-date bone-on-chips through MPS technology, with special emphasis on the scaffold and chip capabilities for mimicking the bone extracellular matrix as this is the key environment generated for cell crosstalk and interaction. The relevant bone models are studied with critical analysis of the methods employed, aiming to serve as a tool for designing new and translational approaches. Additionally, the features reported in these state-of-the-art studies will be useful for modeling bone pathophysiology, guiding future improvements in personalized bone models that can accelerate drug discovery and clinical translation.
Collapse
Affiliation(s)
- Francisco Verdugo-Avello
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | | | - Carlos A Villacis-Aguirre
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Jorge R Toledo
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| |
Collapse
|
5
|
Mallya D, Gadre MA, Varadharajan S, Vasanthan KS. 3D bioprinting for the construction of drug testing models-development strategies and regulatory concerns. Front Bioeng Biotechnol 2025; 13:1457872. [PMID: 40028291 PMCID: PMC11868281 DOI: 10.3389/fbioe.2025.1457872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
A drug to be successfully launched in the market requires a significant amount of capital, resources and time, where the unsuccessful results in the last stages lead to catastrophic failure for discovering drugs. This is the very reason which calls for the invention of innovative models that can closely mimic the human in vivo model for producing reliable results. Throughout the innovation line, there has been improvement in the rationale in silico designing but yet there is requirement for in vitro-in vivo correlations. During the evolving of the drug testing models, the 3D models produced by different methods have been proven to produce better results than the traditional 2D models. However, the in vitro fabrications of live tissues are still bottleneck in realizing their complete potential. There is an urgent need for the development of single, standard and simplified in vitro 3D tissue models that can be reliable for investigating the biological and pathological aspects of drug discovery, which is yet to be achieved. The existing pre-clinical models have considerable drawbacks despite being the gold standard in pre-clinical research. The major drawback being the interspecies differences and low reliability on the generated results. This gap could be overcome by the fabrication of bioengineered human disease models for drug screening. The advancement in the fabrication of 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues. In this review, we have discussed on the evolution of preclinical studies, and different models, including mini tissues, spheroids, organoids, bioengineered three dimensional models and organs on chips. Furthermore, we provide details of different disease models fabricated across various organs and their applications. In addition to this, the review also focuses on the limitations and the current prospects of the role of three dimensionally bioprinted models in drug screening and development.
Collapse
Affiliation(s)
- Divya Mallya
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mrunmayi Ashish Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S. Varadharajan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
6
|
Mazzaglia C, Shery Huang YY, Shields JD. Advancing tumor microenvironment and lymphoid tissue research through 3D bioprinting and biofabrication. Adv Drug Deliv Rev 2025; 217:115485. [PMID: 39653084 DOI: 10.1016/j.addr.2024.115485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Cancer progression is significantly influenced by the complex interactions within the tumor microenvironment (TME). Immune cells, in particular, play a critical role by infiltrating tumors from the circulation and surrounding lymphoid tissues in an attempt to control their spread. However, they often fail in this task. Current in vivo and in vitro preclinical models struggle to fully capture these intricate interactions affecting our ability to understand immune evasion and predict drugs behaviour in the clinic. To address this challenge, biofabrication and particularly 3D bioprinting has emerged as a promising tool for modeling both tumors and the immune system. Its ability to incorporate multiple cell types into 3D matrices, enable tissue compartmentalization with high spatial accuracy, and integrate vasculature makes it a valuable approach. Nevertheless, limited research has focused on capturing the complex tumor-immune interplay in vitro. This review highlights the composition and significance of the TME, the architecture and function of lymphoid tissues, and innovative approaches to modeling their interactions in vitro, while proposing the concept of an extended TME.
Collapse
Affiliation(s)
- Corrado Mazzaglia
- The Nanoscience Centre, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Department of Engineering, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Center for Life Nano, and Neuro-Science of Istituto Italiano di Tecnologia (IIT), Rome 00161, Italy.
| | - Yan Yan Shery Huang
- The Nanoscience Centre, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Department of Engineering, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland
| | - Jacqueline D Shields
- Translational Medical Sciences, School of Medicine, University of Nottingham, Biodiscovery Institute, Nottingham, the United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
7
|
Jiang N, Ying G, Yin Y, Guo J, Lozada J, Valdivia Padilla A, Gómez A, Gomes de Melo BA, Lugo Mestre F, Gansevoort M, Palumbo M, Calá N, Garciamendez-Mijares CE, Kim GA, Takayama S, Gerhard-Herman MD, Zhang YS. A closed-loop modular multiorgan-on-chips platform for self-sustaining and tightly controlled oxygenation. Proc Natl Acad Sci U S A 2024; 121:e2413684121. [PMID: 39541351 PMCID: PMC11588096 DOI: 10.1073/pnas.2413684121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
To mimic physiological microenvironments in organ-on-a-chip systems, physiologically relevant parameters are required to precisely access drug metabolism. Oxygen level is a critical microenvironmental parameter to maintain cellular or tissue functions and modulate their behaviors. Current organ-on-a-chip setups are oftentimes subjected to the ambient incubator oxygen level at 21%, which is higher than most if not all physiological oxygen concentrations. Additionally, the physiological oxygen level in each tissue is different ranging from 0.5 to 13%. Here, a closed-loop modular multiorgan-on-chips platform is developed to enable not only real-time monitoring of the oxygen levels but, more importantly, tight control of them in the range of 4 to 20% across each connected microtissue-on-a-chip in the circulatory culture medium. This platform, which consists of microfluidic oxygen scavenger(s), an oxygen generator, a monitoring/controller system, and bioreactor(s), allows for independent, precise upregulation and downregulation of dissolved oxygen in the perfused culture medium to meet the physiological oxygen level in each modular microtissue compartment, as needed. Furthermore, drug studies using the platform demonstrate that the oxygen level affects drug metabolism in the parallelly connected liver, kidney, and arterial vessel microtissues without organ-organ interactions factored in. Overall, this platform can promote the performances of organ-on-a-chip devices in drug screening by providing more physiologically relevant and independently adjustable oxygen microenvironments for desired organ types on a single- or a multiorgan-on-chip(s) configuration.
Collapse
Affiliation(s)
- Nan Jiang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
- Department of Physics, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Guoliang Ying
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Yixia Yin
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Jie Guo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Jorge Lozada
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Alejandra Valdivia Padilla
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Ameyalli Gómez
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Bruna Alice Gomes de Melo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Francisco Lugo Mestre
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Merel Gansevoort
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Marcello Palumbo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Noemi Calá
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Carlos Ezio Garciamendez-Mijares
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Ge-Ah Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA30318
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA30318
| | - Marie Denis Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Yu Shrike Zhang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
- Harvard Stem Cell Institute, Harvard University,Cambridge, MA02138
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA02142
| |
Collapse
|
8
|
Eliahoo P, Setayesh H, Hoffman T, Wu Y, Li S, Treweek JB. Viscoelasticity in 3D Cell Culture and Regenerative Medicine: Measurement Techniques and Biological Relevance. ACS MATERIALS AU 2024; 4:354-384. [PMID: 39006396 PMCID: PMC11240420 DOI: 10.1021/acsmaterialsau.3c00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 07/16/2024]
Abstract
The field of mechanobiology is gaining prominence due to recent findings that show cells sense and respond to the mechanical properties of their environment through a process called mechanotransduction. The mechanical properties of cells, cell organelles, and the extracellular matrix are understood to be viscoelastic. Various technologies have been researched and developed for measuring the viscoelasticity of biological materials, which may provide insight into both the cellular mechanisms and the biological functions of mechanotransduction. Here, we explain the concept of viscoelasticity and introduce the major techniques that have been used to measure the viscoelasticity of various soft materials in different length- and timescale frames. The topology of the material undergoing testing, the geometry of the probe, the magnitude of the exerted stress, and the resulting deformation should be carefully considered to choose a proper technique for each application. Lastly, we discuss several applications of viscoelasticity in 3D cell culture and tissue models for regenerative medicine, including organoids, organ-on-a-chip systems, engineered tissue constructs, and tunable viscoelastic hydrogels for 3D bioprinting and cell-based therapies.
Collapse
Affiliation(s)
- Payam Eliahoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089 United States
| | - Hesam Setayesh
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089 United States
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Yifan Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Jennifer B Treweek
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089 United States
| |
Collapse
|
9
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
10
|
De Vitis E, Stanzione A, Romano A, Quattrini A, Gigli G, Moroni L, Gervaso F, Polini A. The Evolution of Technology-Driven In Vitro Models for Neurodegenerative Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304989. [PMID: 38366798 DOI: 10.1002/advs.202304989] [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: 07/21/2023] [Revised: 01/15/2024] [Indexed: 02/18/2024]
Abstract
The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
Collapse
Affiliation(s)
- Eleonora De Vitis
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Antonella Stanzione
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Angelo Quattrini
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Giuseppe Gigli
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Lorenzo Moroni
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, Netherlands
| | - Francesca Gervaso
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
11
|
Balachander GM, Nilawar S, Meka SRK, Ghosh LD, Chatterjee K. Unravelling microRNA regulation and miRNA-mRNA regulatory networks in osteogenesis driven by 3D nanotopographical cues. Biomater Sci 2024; 12:978-989. [PMID: 38189225 DOI: 10.1039/d3bm01597a] [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: 01/09/2024]
Abstract
Three-dimensional (3D) culturing of cells is being adopted for developing tissues for various applications such as mechanistic studies, drug testing, tissue regeneration, and animal-free meat. These approaches often involve cost-effective differentiation of stem or progenitor cells. One approach is to exploit architectural cues on a 3D substrate to drive cellular differentiation, which has been shown to be effective in various studies. Although extensive gene expression data from such studies have shown that gene expression patterns might differ, the gene regulatory networks controlling the expression of genes are rarely studied. In this study, we profiled genes and microRNAs (miRNAs) via next-generation sequencing (NGS) in human mesenchymal stem cells (hMSCs) driven toward osteogenesis via architectural cues in 3D matrices (3D conditions) and compared with cells in two-dimensional (2D) culture driven toward osteogenesis via soluble osteoinductive factors (OF conditions). The total number of differentially expressed genes was smaller in 3D compared to OF conditions. A distinct set of genes was observed under these conditions that have been shown to control osteogenic differentiation via different pathways. Small RNA sequencing revealed a core set of miRNAs to be differentially expressed under these conditions, similar to those that have been previously implicated in osteogenesis. We also observed a distinct regulation of miRNAs in these samples that can modulate gene expression, suggesting supplementary gene regulatory networks operative under different stimuli. This study provides insights into studying gene regulatory networks for identifying critical nodes to target for enhanced cellular differentiation and reveal the differences in physical and biochemical cues to drive cell fates.
Collapse
Affiliation(s)
- Gowri Manohari Balachander
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Sagar Nilawar
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| | - Sai Rama Krishna Meka
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| | - Lopamudra Das Ghosh
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| |
Collapse
|
12
|
Hahn F, Ferrandez-Montero A, Queri M, Vancaeyzeele C, Plesse C, Agniel R, Leroy-Dudal J. Electroactive 4D Porous Scaffold Based on Conducting Polymer as a Responsive and Dynamic In Vitro Cell Culture Platform. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5613-5626. [PMID: 38278772 PMCID: PMC10859895 DOI: 10.1021/acsami.3c16686] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
In vivo, cells reside in a 3D porous and dynamic microenvironment. It provides biochemical and biophysical cues that regulate cell behavior in physiological and pathological processes. In the context of fundamental cell biology research, tissue engineering, and cell-based drug screening systems, a challenge is to develop relevant in vitro models that could integrate the dynamic properties of the cell microenvironment. Taking advantage of the promising high internal phase emulsion templating, we here designed a polyHIPE scaffold with a wide interconnected porosity and functionalized its internal 3D surface with a thin layer of electroactive conducting polymer poly(3,4-ethylenedioxythiophene) (PEDOT) to turn it into a 4D electroresponsive scaffold. The resulting scaffold was cytocompatible with fibroblasts, supported cellular infiltration, and hosted cells, which display a 3D spreading morphology. It demonstrated robust actuation in ion- and protein-rich complex culture media, and its electroresponsiveness was not altered by fibroblast colonization. Thanks to customized electrochemical stimulation setups, the electromechanical response of the polyHIPE/PEDOT scaffolds was characterized in situ under a confocal microscope and showed 10% reversible volume variations. Finally, the setups were used to monitor in real time and in situ fibroblasts cultured into the polyHIPE/PEDOT scaffold during several cycles of electromechanical stimuli. Thus, we demonstrated the proof of concept of this tunable scaffold as a tool for future 4D cell culture and mechanobiology studies.
Collapse
Affiliation(s)
- Franziska Hahn
- Equipe
de Recherche sur les Relations Matrice Extracellulaire-Cellules (ERRMECe),
Groupe Matrice Extracellulaire et Physiopathologie (MECuP), I-Mat, CY Cergy Paris Université, 95000 Neuville
sur Oise, France
- Laboratoire
de Physicochimie des Polymères et des Interfaces (LPPI), I-Mat, CY Cergy Paris Université, 95000 Neuville sur Oise, France
| | - Ana Ferrandez-Montero
- Equipe
de Recherche sur les Relations Matrice Extracellulaire-Cellules (ERRMECe),
Groupe Matrice Extracellulaire et Physiopathologie (MECuP), I-Mat, CY Cergy Paris Université, 95000 Neuville
sur Oise, France
- Laboratoire
de Physicochimie des Polymères et des Interfaces (LPPI), I-Mat, CY Cergy Paris Université, 95000 Neuville sur Oise, France
- Instituto
de Ceramica y Vidrio (ICV), CSIC, Campus Cantoblanco, Kelsen 5., 28049 Madrid, Spain
| | - Mélodie Queri
- Equipe
de Recherche sur les Relations Matrice Extracellulaire-Cellules (ERRMECe),
Groupe Matrice Extracellulaire et Physiopathologie (MECuP), I-Mat, CY Cergy Paris Université, 95000 Neuville
sur Oise, France
- Laboratoire
de Physicochimie des Polymères et des Interfaces (LPPI), I-Mat, CY Cergy Paris Université, 95000 Neuville sur Oise, France
| | - Cédric Vancaeyzeele
- Laboratoire
de Physicochimie des Polymères et des Interfaces (LPPI), I-Mat, CY Cergy Paris Université, 95000 Neuville sur Oise, France
| | - Cédric Plesse
- Laboratoire
de Physicochimie des Polymères et des Interfaces (LPPI), I-Mat, CY Cergy Paris Université, 95000 Neuville sur Oise, France
| | - Rémy Agniel
- Equipe
de Recherche sur les Relations Matrice Extracellulaire-Cellules (ERRMECe),
Groupe Matrice Extracellulaire et Physiopathologie (MECuP), I-Mat, CY Cergy Paris Université, 95000 Neuville
sur Oise, France
| | - Johanne Leroy-Dudal
- Equipe
de Recherche sur les Relations Matrice Extracellulaire-Cellules (ERRMECe),
Groupe Matrice Extracellulaire et Physiopathologie (MECuP), I-Mat, CY Cergy Paris Université, 95000 Neuville
sur Oise, France
| |
Collapse
|
13
|
Zehorai E, Maor-Shoshani A, Molotski N, Dorojkin A, Marelly N, Dvash T, Lavon N. From fertilised oocyte to cultivated meat - harnessing bovine embryonic stem cells in the cultivated meat industry. Reprod Fertil Dev 2023; 36:124-132. [PMID: 38064188 DOI: 10.1071/rd23169] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Global demand for animal protein is on the rise, but many practices common in conventional production are no longer scalable due to environmental impact, public health concerns, and fragility of food systems. For these reasons and more, a pressing need has arisen for sustainable, nutritious, and animal welfare-conscious sources of protein, spurring research dedicated to the production of cultivated meat. Meat mainly consists of muscle, fat, and connective tissue, all of which can be sourced and differentiated from pluripotent stem cells to resemble their nutritional values in muscle tissue. In this paper, we outline the approach that we took to derive bovine embryonic stem cell lines (bESCs) and to characterise them using FACS (fluorescence-activated cell sorting), real-time PCR and immunofluorescence staining. We show their cell growth profile and genetic stability and demonstrate their induced differentiation to mesoderm committed cells. In addition, we discuss our strategy for preparation of master and working cell banks, by which we can expand and grow cells in suspension in quantities suitable for mass production. Consequently, we demonstrate the potential benefits of harnessing bESCs in the production of cultivated meat.
Collapse
Affiliation(s)
| | | | | | | | | | - Tami Dvash
- Aleph Farms Ltd, Rehovot 7670401, Israel
| | - Neta Lavon
- Aleph Farms Ltd, Rehovot 7670401, Israel
| |
Collapse
|
14
|
Sabzevari A, Rayat Pisheh H, Ansari M, Salati A. Progress in bioprinting technology for tissue regeneration. J Artif Organs 2023; 26:255-274. [PMID: 37119315 DOI: 10.1007/s10047-023-01394-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/09/2023] [Indexed: 05/01/2023]
Abstract
In recent years, due to the increase in diseases that require organ/tissue transplantation and the limited donor, on the other hand, patients have lost hope of recovery and organ transplantation. Regenerative medicine is one of the new sciences that promises a bright future for these patients by providing solutions to repair, improve function, and replace tissue. One of the technologies used in regenerative medicine is three-dimensional (3D) bioprinters. Bioprinting is a new strategy that is the basis for starting a global revolution in the field of medical sciences and has attracted much attention. 3D bioprinters use a combination of advanced biology and cell science, computer science, and materials science to create complex bio-hybrid structures for various applications. The capacity to use this technology can be demonstrated in regenerative medicine to make various connective tissues, such as skin, cartilage, and bone. One of the essential parts of a 3D bioprinter is the bio-ink. Bio-ink is a combination of biologically active molecules, cells, and biomaterials that make the printed product. In this review, we examine the main bioprinting strategies, such as inkjet printing, laser, and extrusion-based bioprinting, as well as some of their applications.
Collapse
Affiliation(s)
- Alireza Sabzevari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | | | - Mojtaba Ansari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran.
| | - Amir Salati
- Tissue Engineering and Applied Cell Sciences Group, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
15
|
Leng Y, Li X, Zheng F, Liu H, Wang C, Wang X, Liao Y, Liu J, Meng K, Yu J, Zhang J, Wang B, Tan Y, Liu M, Jia X, Li D, Li Y, Gu Z, Fan Y. Advances in In Vitro Models of Neuromuscular Junction: Focusing on Organ-on-a-Chip, Organoids, and Biohybrid Robotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211059. [PMID: 36934404 DOI: 10.1002/adma.202211059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/18/2023] [Indexed: 06/18/2023]
Abstract
The neuromuscular junction (NMJ) is a peripheral synaptic connection between presynaptic motor neurons and postsynaptic skeletal muscle fibers that enables muscle contraction and voluntary motor movement. Many traumatic, neurodegenerative, and neuroimmunological diseases are classically believed to mainly affect either the neuronal or the muscle side of the NMJ, and treatment options are lacking. Recent advances in novel techniques have helped develop in vitro physiological and pathophysiological models of the NMJ as well as enable precise control and evaluation of its functions. This paper reviews the recent developments in in vitro NMJ models with 2D or 3D cultures, from organ-on-a-chip and organoids to biohybrid robotics. Related derivative techniques are introduced for functional analysis of the NMJ, such as the patch-clamp technique, microelectrode arrays, calcium imaging, and stimulus methods, particularly optogenetic-mediated light stimulation, microelectrode-mediated electrical stimulation, and biochemical stimulation. Finally, the applications of the in vitro NMJ models as disease models or for drug screening related to suitable neuromuscular diseases are summarized and their future development trends and challenges are discussed.
Collapse
Affiliation(s)
- Yubing Leng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Xiaorui Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, 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
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xudong Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jiangyue Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Kaiqi Meng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jiaheng Yu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jingyi Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Binyu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Meili Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Xiaoling Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Deyu Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| |
Collapse
|
16
|
Huskin G, Chen J, Davis T, Jun HW. Tissue-Engineered 3D In Vitro Disease Models for High-Throughput Drug Screening. Tissue Eng Regen Med 2023; 20:523-538. [PMID: 36892736 PMCID: PMC10313592 DOI: 10.1007/s13770-023-00522-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 03/10/2023] Open
Abstract
During high-throughput drug screening, in vitro models are fabricated and the effects of therapeutics on the models evaluated in high throughput-for example, with automated liquid handling systems and microplate reader-based high-throughput screening (HTS) assays. The most frequently-used model systems for HTS, 2D models, do not adequately model the in vivo 3D microenvironment-an important aspect of which is the extracellular matrix-and therefore, 2D models may not be appropriate for drug screening. Instead, tissue-engineered 3D models with extracellular matrix-mimicking components are destined to become the preferred in vitro systems for HTS. However, for 3D models, such as 3D cell-laden hydrogels and scaffolds, cell sheets, and spheroids as well as 3D microfluidic and organ-on-a-chip systems, to replace 2D models in HTS, they must be compatible with high-throughput fabrication schemes and evaluation methods. In this review, we summarize HTS in 2D models and discuss recent studies that have successfully demonstrated HTS-compatible 3D models of high-impact diseases, such as cancers or cardiovascular diseases.
Collapse
Affiliation(s)
- Gillian Huskin
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jun Chen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Trenton Davis
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
17
|
Doganay MT, Chelliah CJ, Tozluyurt A, Hujer AM, Obaro SK, Gurkan U, Patel R, Bonomo RA, Draz M. 3D Printed Materials for Combating Antimicrobial Resistance. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2023; 67:371-398. [PMID: 37790286 PMCID: PMC10545363 DOI: 10.1016/j.mattod.2023.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Three-dimensional (3D) printing is a rapidly growing technology with a significant capacity for translational applications in both biology and medicine. 3D-printed living and non-living materials are being widely tested as a potential replacement for conventional solutions for testing and combating antimicrobial resistance (AMR). The precise control of cells and their microenvironment, while simulating the complexity and dynamics of an in vivo environment, provides an excellent opportunity to advance the modeling and treatment of challenging infections and other health conditions. 3D-printing models the complicated niches of microbes and host-pathogen interactions, and most importantly, how microbes develop resistance to antibiotics. In addition, 3D-printed materials can be applied to testing and delivering antibiotics. Here, we provide an overview of 3D printed materials and biosystems and their biomedical applications, focusing on ever increasing AMR. Recent applications of 3D printing to alleviate the impact of AMR, including developed bioprinted systems, targeted bacterial infections, and tested antibiotics are presented.
Collapse
Affiliation(s)
- Mert Tunca Doganay
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Cyril John Chelliah
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Abdullah Tozluyurt
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Andrea M Hujer
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | | | - Umut Gurkan
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology and Division of Public Health, Infectious Diseases, and Occupational medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert A Bonomo
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES) Cleveland, OH, USA
| | - Mohamed Draz
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
18
|
Krishnakumar A, Kadian S, Heredia Rivera U, Chittiboyina S, Lelièvre SA, Rahimi R. Organ-on-a-Chip Platform with an Integrated Screen-Printed Electrode Array for Real-Time Monitoring Trans-Epithelial Barrier and Bubble Formation. ACS Biomater Sci Eng 2023; 9:1620-1628. [PMID: 36763005 DOI: 10.1021/acsbiomaterials.2c00494] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Cellular tight junctions play a key role in establishing a barrier between different compartments of the body by regulating the selective passage of different solutes across epithelial and endothelial tissues. Over the past decade, significant efforts have been conducted to develop more clinically relevant "organ-on-a-chip" models with integrated trans-epithelial electrical resistance (TEER) monitoring systems to help better understand the fundamental underpinnings of epithelial tissue physiology upon exposure to different substances. However, most of these platforms require the use of high-cost and time-consuming photolithography processes, which limits their scalability and practical implementation in clinical research. To address this need, we have developed a low-cost microfluidic platform with an integrated electrode array that allows continuous real-time monitoring of TEER and the risk of bubble formation in the microfluidic system by using scalable manufacturing technologies such as screen printing and laser processing. The integrated printed electrode array exhibited excellent stability (with less than ∼0.02 Ω change in resistance) even after long-term exposure to a complex culture medium. As a proof of concept, the fully integrated platform was tested with HMT3522 S1 epithelial cells to evaluate the tight barrier junction formation through TEER measurement and validated with standard immunostaining procedures for Zonula occludens-1 protein. This platform could be regarded as a stepping stone for the fabrication of disposable and low-cost organ and tissue-on-a-chip models with integrated sensors to facilitate studying the dynamic response of epithelial tissues to different substances in more physiologically relevant conditions.
Collapse
Affiliation(s)
- Akshay Krishnakumar
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sachin Kadian
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
- School of Materials Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ulisses Heredia Rivera
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
- School of Materials Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shirisha Chittiboyina
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sophie A Lelièvre
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana 47907, United States
| | - Rahim Rahimi
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
- School of Materials Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
19
|
Hwang KS, Seo EU, Choi N, Kim J, Kim HN. 3D engineered tissue models for studying human-specific infectious viral diseases. Bioact Mater 2023; 21:576-594. [PMID: 36204281 PMCID: PMC9519398 DOI: 10.1016/j.bioactmat.2022.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/13/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Viral infections cause damage to various organ systems by inducing organ-specific symptoms or systemic multi-organ damage. Depending on the infection route and virus type, infectious diseases are classified as respiratory, nervous, immune, digestive, or skin infections. Since these infectious diseases can widely spread in the community and their catastrophic effects are severe, identification of their causative agent and mechanisms underlying their pathogenesis is an urgent necessity. Although infection-associated mechanisms have been studied in two-dimensional (2D) cell culture models and animal models, they have shown limitations in organ-specific or human-associated pathogenesis, and the development of a human-organ-mimetic system is required. Recently, three-dimensional (3D) engineered tissue models, which can present human organ-like physiology in terms of the 3D structure, utilization of human-originated cells, recapitulation of physiological stimuli, and tight cell–cell interactions, were developed. Furthermore, recent studies have shown that these models can recapitulate infection-associated pathologies. In this review, we summarized the recent advances in 3D engineered tissue models that mimic organ-specific viral infections. First, we briefly described the limitations of the current 2D and animal models in recapitulating human-specific viral infection pathology. Next, we provided an overview of recently reported viral infection models, focusing particularly on organ-specific infection pathologies. Finally, a future perspective that must be pursued to reconstitute more human-specific infectious diseases is presented. 3D in vitro models are different from the traditional model in the infection process. Human-specific infection research requires a 3D microenvironment and human cells. 3D in vitro infectious models can be useful for basic research on infectious disease. 3D in vitro infectious models recapitulate the complex cell-virus-immune interaction.
Collapse
Affiliation(s)
- Kyeong Seob Hwang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eun U Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Jongbaeg Kim
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Corresponding author.
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- Corresponding author. Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
20
|
Plug-and-Play Lymph Node-on-Chip: Secondary Tumor Modeling by the Combination of Cell Spheroid, Collagen Sponge and T-Cells. Int J Mol Sci 2023; 24:ijms24043183. [PMID: 36834594 PMCID: PMC9966643 DOI: 10.3390/ijms24043183] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/08/2023] Open
Abstract
Towards the improvement of the efficient study of drugs and contrast agents, the 3D microfluidic platforms are currently being actively developed for testing these substances and particles in vitro. Here, we have elaborated a microfluidic lymph node-on-chip (LNOC) as a tissue engineered model of a secondary tumor in lymph node (LN) formed due to the metastasis process. The developed chip has a collagen sponge with a 3D spheroid of 4T1 cells located inside, simulating secondary tumor in the lymphoid tissue. This collagen sponge has a morphology and porosity comparable to that of a native human LN. To demonstrate the suitability of the obtained chip for pharmacological applications, we used it to evaluate the effect of contrast agent/drug carrier size, on the penetration and accumulation of particles in 3D spheroids modeling secondary tumor. For this, the 0.3, 0.5 and 4 μm bovine serum albumin (BSA)/tannic acid (TA) capsules were mixed with lymphocytes and pumped through the developed chip. The capsule penetration was examined by scanning with fluorescence microscopy followed by quantitative image analysis. The results show that capsules with a size of 0.3 μm passed more easily to the tumor spheroid and penetrated inside. We hope that the device will represent a reliable alternative to in vivo early secondary tumor models and decrease the amount of in vivo experiments in the frame of preclinical study.
Collapse
|
21
|
Takahashi H, Wakayama H, Nagase K, Shimizu T. Engineered Human Muscle Tissue from Multilayered Aligned Myofiber Sheets for Studies of Muscle Physiology and Predicting Drug Response. SMALL METHODS 2023; 7:e2200849. [PMID: 36562139 DOI: 10.1002/smtd.202200849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/22/2022] [Indexed: 06/17/2023]
Abstract
In preclinical drug testing, human muscle tissue models are critical to understanding the complex physiology, including drug effects in the human body. This study reports that a multilayering approach to cell sheet-based engineering produces an engineered human muscle tissue with sufficient contractile force suitable for measurement. A thermoresponsive micropatterned substrate regulates the biomimetic alignment of myofiber structures enabling the harvest of the aligned myofibers as a single cell sheet. The functional muscle tissue is produced by layering multiple myofiber sheets on a fibrin-based gel. This gel environment promotes myofiber maturation, provides the tissue an elastic platform for contraction, and allows the attachment of a measurement device. Since this multilayering approach is effective in enhancing the contractile ability of the muscle tissue, this muscle tissue generates a significantly high contractile force that can be measured quantitatively. The multilayered muscle tissue shows unidirectional contraction from electrical and chemical stimulation. In addition, their physiological responses to representative drugs can be determined quantitatively in real time by changes in contractile force and fatigue resistance. These physiological properties indicate that the engineered muscle tissue can become a promising tissue model for preclinical in vitro studies in muscle physiology and drug discovery.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Haruno Wakayama
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| |
Collapse
|
22
|
He S, Lim GE. The Application of High-Throughput Approaches in Identifying Novel Therapeutic Targets and Agents to Treat Diabetes. Adv Biol (Weinh) 2023; 7:e2200151. [PMID: 36398493 DOI: 10.1002/adbi.202200151] [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: 05/31/2022] [Revised: 10/04/2022] [Indexed: 11/19/2022]
Abstract
During the past decades, unprecedented progress in technologies has revolutionized traditional research methodologies. Among these, advances in high-throughput drug screening approaches have permitted the rapid identification of potential therapeutic agents from drug libraries that contain thousands or millions of molecules. Moreover, high-throughput-based therapeutic target discovery strategies can comprehensively interrogate relationships between biomolecules (e.g., gene, RNA, and protein) and diseases and significantly increase the authors' knowledge of disease mechanisms. Diabetes is a chronic disease primarily characterized by the incapacity of the body to maintain normoglycemia. The prevalence of diabetes in modern society has become a severe public health issue that threatens the well-being of millions of patients. Although a number of pharmacological treatments are available, there is no permanent cure for diabetes, and discovering novel therapeutic targets and agents continues to be an urgent need. The present review discusses the technical details of high-throughput screening approaches in drug discovery, followed by introducing the applications of such approaches to diabetes research. This review aims to provide an example of the applicability of high-throughput technologies in facilitating different aspects of disease research.
Collapse
Affiliation(s)
- Siyi He
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| |
Collapse
|
23
|
Assad H, Assad A, Kumar A. Recent Developments in 3D Bio-Printing and Its Biomedical Applications. Pharmaceutics 2023; 15:255. [PMID: 36678884 PMCID: PMC9861443 DOI: 10.3390/pharmaceutics15010255] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The fast-developing field of 3D bio-printing has been extensively used to improve the usability and performance of scaffolds filled with cells. Over the last few decades, a variety of tissues and organs including skin, blood vessels, and hearts, etc., have all been produced in large quantities via 3D bio-printing. These tissues and organs are not only able to serve as building blocks for the ultimate goal of repair and regeneration, but they can also be utilized as in vitro models for pharmacokinetics, drug screening, and other purposes. To further 3D-printing uses in tissue engineering, research on novel, suitable biomaterials with quick cross-linking capabilities is a prerequisite. A wider variety of acceptable 3D-printed materials are still needed, as well as better printing resolution (particularly at the nanoscale range), speed, and biomaterial compatibility. The aim of this study is to provide expertise in the most prevalent and new biomaterials used in 3D bio-printing as well as an introduction to the associated approaches that are frequently considered by researchers. Furthermore, an effort has been made to convey the most pertinent implementations of 3D bio-printing processes, such as tissue regeneration, etc., by providing the most significant research together with a comprehensive list of material selection guidelines, constraints, and future prospects.
Collapse
Affiliation(s)
- Humira Assad
- Department of Chemistry, School of Chemical Engineering and Physical Sciences, Lovely Professional University, Punjab 144001, India
| | - Arvina Assad
- Bibi Halima College of Nursing and Medical Technology, Srinagar 190010, India
| | - Ashish Kumar
- Nalanda College of Engineering, Department of Science and Technology, Government of Bihar, Patna 803108, India
| |
Collapse
|
24
|
Iqbal MJ, Javed Z, Herrera-Bravo J, Sadia H, Anum F, Raza S, Tahir A, Shahwani MN, Sharifi-Rad J, Calina D, Cho WC. Biosensing chips for cancer diagnosis and treatment: a new wave towards clinical innovation. Cancer Cell Int 2022; 22:354. [PMCID: PMC9664821 DOI: 10.1186/s12935-022-02777-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractRecent technological advances in nanoscience and material designing have led to the development of point-of-care devices for biomolecule sensing and cancer diagnosis. In situ and portable sensing devices for bedside, diagnosis can effectively improve the patient’s clinical outcomes and reduce the mortality rate. Detection of exosomal RNAs by immuno-biochip with increased sensitivity and specificity to diagnose cancer has raised the understanding of the tumor microenvironment and many other technology-based biosensing devices hold great promise for clinical innovations to conquer the unbeatable fort of cancer metastasis. Electrochemical biosensors are the most sensitive category of biomolecule detection sensors with significantly low concentrations down to the atomic level. In this sense, this review addresses the recent advances in cancer detection and diagnosis by developing significant biological sensing devices that are believed to have better sensing potential than existing facilities.
Collapse
|
25
|
Chiang MC, Nicol CJB, Lo SS, Hung SW, Wang CJ, Lin CH. Resveratrol Mitigates Oxygen and Glucose Deprivation-Induced Inflammation, NLRP3 Inflammasome, and Oxidative Stress in 3D Neuronal Culture. Int J Mol Sci 2022; 23:ijms231911678. [PMID: 36232980 PMCID: PMC9570351 DOI: 10.3390/ijms231911678] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Oxygen glucose deprivation (OGD) can produce hypoxia-induced neurotoxicity and is a mature in vitro model of hypoxic cell damage. Activated AMP-activated protein kinase (AMPK) regulates a downstream pathway that substantially increases bioenergy production, which may be a key player in physiological energy and has also been shown to play a role in regulating neuroprotective processes. Resveratrol is an effective activator of AMPK, indicating that it may have therapeutic potential as a neuroprotective agent. However, the mechanism by which resveratrol achieves these beneficial effects in SH-SY5Y cells exposed to OGD-induced inflammation and oxidative stress in a 3D gelatin scaffold remains unclear. Therefore, in the present study, we investigated the effect of resveratrol in 3D gelatin scaffold cells to understand its neuroprotective effects on NF-κB signaling, NLRP3 inflammasome, and oxidative stress under OGD conditions. Here, we show that resveratrol improves the expression levels of cell viability, inflammatory cytokines (TNF-α, IL-1β, and IL-18), NF-κB signaling, and NLRP3 inflammasome, that OGD increases. In addition, resveratrol rescued oxidative stress, nuclear factor-erythroid 2 related factor 2 (Nrf2), and Nrf2 downstream antioxidant target genes (e.g., SOD, Gpx GSH, catalase, and HO-1). Treatment with resveratrol can significantly normalize OGD-induced changes in SH-SY5Y cell inflammation, oxidative stress, and oxidative defense gene expression; however, these resveratrol protective effects are affected by AMPK antagonists (Compounds C) blocking. These findings improve our understanding of the mechanism of the AMPK-dependent protective effect of resveratrol under 3D OGD-induced inflammation and oxidative stress-mediated cerebral ischemic stroke conditions.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei 242304, Taiwan
| | - Christopher J. B. Nicol
- Departments of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
- Departments of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
- Cancer Biology and Genetics Division, Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Shy-Shyong Lo
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei 242304, Taiwan
| | - Shiang-Wei Hung
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei 242304, Taiwan
| | - Chieh-Ju Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei 242304, Taiwan
| | - Chien-Hung Lin
- Division of Pediatric Immunology and Nephrology, Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Pediatrics, Zhongxing Branch, Taipei City Hospital, Taipei 10341, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- College of Science and Engineering, Fu Jen Catholic University, New Taipei 242304, Taiwan
- Correspondence:
| |
Collapse
|
26
|
Ng WL, Win Naing M, Suntornnond R, Vijayavenkataraman S. Editorial: Fabrication of in-vitro 3D human tissue models—From cell processing to advanced manufacturing. Front Bioeng Biotechnol 2022; 10:1035601. [PMID: 36225605 PMCID: PMC9549280 DOI: 10.3389/fbioe.2022.1035601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Wei Long Ng
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University (NTU), Singapore, Singapore
- *Correspondence: Wei Long Ng,
| | - May Win Naing
- Singapore Institute of Manufacturing Technology (SIMTech), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ratima Suntornnond
- Biomanufacturing Technology, Bioprocessing Technology Institute (BTI), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Sanjairaj Vijayavenkataraman
- The Vijay Lab, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, United States
| |
Collapse
|
27
|
Takahashi H, Yoshida A, Gao B, Yamanaka K, Shimizu T. Harvest of quality-controlled bovine myogenic cells and biomimetic bovine muscle tissue engineering for sustainable meat production. Biomaterials 2022; 287:121649. [PMID: 35779482 DOI: 10.1016/j.biomaterials.2022.121649] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 05/19/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022]
Abstract
Alternative technology for meat production holds the potential to alleviate ethical, environmental, and public health concerns associated with conventional meat production. Cultured meat produced using cell culture technology promises to become a viable alternative to animal-raised meat for the future of the food industry. In this study, biomimetic bovine muscle tissue was artificially fabricated from myogenic cells extracted from bovine meat. Our primary culture method relies on three key factors; a sequential digesting process, enzymatic treatment with pronase, and coating with laminin fragment on culture dishes. This method allows the efficient collection of large numbers of primary cells from bovine cheek meat, purifies the myogenic cells from the cell mixture, and then continuously grows the myogenic cells in vitro. In addition, using our "quality control" methods, we were able to determine the "cell quality", including the proliferative and differentiation capability in each step of the primary culture. Furthermore, to mimic native bovine meat, the quality-controlled bovine myogenic cells were cultured on a micropatterned thermoresponsive substrate stimulating a native-like aligned structure of cells, which were then transferred onto a fibrin-based gel. This gel-based culture environment promoted structural and functional maturation of the myogenic cells, resulting in the production of bovine muscle tissues with sarcomere structures, native-like membrane structures, and contractile ability. We believe that these biomimetic features of "tissue-engineered meat" are important for the production of future cultured meat, which will need native-like nutrients, texture and taste. Therefore, our meat production approach will provide a new platform to produce more native biomimetic tissue-engineered meat in the near future.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan.
| | - Azumi Yoshida
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| | - Botao Gao
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| | - Kumiko Yamanaka
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| |
Collapse
|
28
|
Additive manufacturing in respiratory sciences - Current applications and future prospects. Adv Drug Deliv Rev 2022; 186:114341. [PMID: 35569558 DOI: 10.1016/j.addr.2022.114341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/21/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022]
Abstract
Additive Manufacturing (AM) comprises a variety of techniques that enable fabrication of customised objects with specific attributes. The versatility of AM procedures and constant technological improvements allow for their application in the development of medicinal products and medical devices. This review provides an overview of AM applications related to respiratory sciences. For this purpose, both fields of research are briefly introduced and the potential benefits of integrating AM to respiratory sciences at different levels of pharmaceutical development are highlighted. Tailored manufacturing of microstructures as a particle design approach in respiratory drug delivery will be discussed. At the dosage form level, we exemplify AM as an important link in the iterative loop of data driven inhaler design, rapid prototyping and in vitro testing. This review also presents the application of bioprinting in the respiratory field for design of biorelevant in vitro cellular models, followed by an overview of AM-related processes in preventive and therapeutic care. Finally, this review discusses future prospects of AM as a component in a digital health environment.
Collapse
|
29
|
Hammel JH, Zatorski JM, Cook SR, Pompano RR, Munson JM. Engineering in vitro immune-competent tissue models for testing and evaluation of therapeutics. Adv Drug Deliv Rev 2022; 182:114111. [PMID: 35031388 PMCID: PMC8908413 DOI: 10.1016/j.addr.2022.114111] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/16/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Advances in 3D cell culture, microscale fluidic control, and cellular analysis have enabled the development of more physiologically-relevant engineered models of human organs with precise control of the cellular microenvironment. Engineered models have been used successfully to answer fundamental biological questions and to screen therapeutics, but these often neglect key elements of the immune system. There are immune elements in every tissue that contribute to healthy and diseased states. Including immune function will be essential for effective preclinical testing of therapeutics for inflammatory and immune-modulated diseases. In this review, we first discuss the key components to consider in designing engineered immune-competent models in terms of physical, chemical, and biological cues. Next, we review recent applications of models of immunity for screening therapeutics for cancer, preclinical evaluation of engineered T cells, modeling autoimmunity, and screening vaccine efficacy. Future work is needed to further recapitulate immune responses in engineered models for the most informative therapeutic screening and evaluation.
Collapse
Affiliation(s)
- Jennifer H. Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| | - Jonathan M. Zatorski
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Sophie R. Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA,Department of Biomedical Engineering, University of Virginia; Charlottesville, Virginia 22904, USA,Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| |
Collapse
|
30
|
Rabie AM, Ali ASM, Al-Zeer MA, Barhoum A, EL-Hallouty S, Shousha WG, Berg J, Kurreck J, Khalil ASG. Spontaneous Formation of 3D Breast Cancer Tissues on Electrospun Chitosan/Poly(ethylene oxide) Nanofibrous Scaffolds. ACS OMEGA 2022; 7:2114-2126. [PMID: 35071900 PMCID: PMC8771982 DOI: 10.1021/acsomega.1c05646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/22/2021] [Indexed: 05/06/2023]
Abstract
Three-dimensional (3D) tissue culture has attracted a great deal of attention as a result of the need to replace the conventional two-dimensional cell cultures with more meaningful methods, especially for understanding the sophisticated nature of native tumor microenvironments. However, most techniques for 3D tissue culture are laborious, expensive, and limited to spheroid formation. In this study, a low-cost and highly effective nanofibrous scaffold is presented for spontaneous formation of reproducible 3D breast cancer microtissues. Experimentally, aligned and non-aligned chitosan/poly(ethylene oxide) nanofibrous scaffolds were prepared at one of two chitosan concentrations (2 and 4 wt %) and various electrospinning parameters. The resulting fabricated scaffolds (C2P1 and C4P1) were structurally and morphologically characterized, as well as analyzed in silico. The obtained data suggest that the fiber diameter, surface roughness, and scaffold wettability are tunable and can be influenced based on the chitosan concentration, electrospinning conditions, and alignment mode. To test the usefulness of the fabricated scaffolds for 3D cell culture, a breast cancer cell line (MCF-7) was cultured on their surfaces and evaluated morphologically and biochemically. The obtained data showed a higher proliferation rate for cells grown on scaffolds compared to cells grown on two-dimensional adherent plates (tissue culture plate). The MTT assay revealed that the rate of cell proliferation on nanofibrous scaffolds is statistically significantly higher compared to tissue culture plate (P ≤ 0.001) after 14 days of culture. The formation of spheroids within the first few days of culture shows that the scaffolds effectively support 3D tissue culture from the outset of the experiment. Furthermore, 3D breast cancer tissues were spontaneously formed within 10 days of culture on aligned and non-aligned nanofibrous scaffolds, which suggests that the scaffolds imitate the in vivo extracellular matrix in the tumor microenvironment. Detailed mechanisms for the spontaneous formation of the 3D microtissues have been proposed. Our results suggest that scaffold surface topography significantly influences tissue formation and behavior of the cells.
Collapse
Affiliation(s)
- Amna M.
I. Rabie
- Environmental
and Smart Technology Group (ESTG), Faculty of Science, Fayoum University, 63514 Fayoum, Egypt
- Chemistry
Department, Faculty of Science, Helwan University, Ain Helwan, 11795 Cairo, Egypt
| | - Ahmed S. M. Ali
- Department
of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
- Nanotechnology
Research Center (NTRC), The British University
in Egypt (BUE), El-Sherouk City, 11837 Cairo, Egypt
| | - Munir A. Al-Zeer
- Department
of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Ahmed Barhoum
- Chemistry
Department, Faculty of Science, Helwan University, Ain Helwan, 11795 Cairo, Egypt
| | - Salwa EL-Hallouty
- Department
of Medicinal Drugs, National Research Center, 12622 Giza, Egypt
| | - Wafaa G. Shousha
- Chemistry
Department, Faculty of Science, Helwan University, Ain Helwan, 11795 Cairo, Egypt
| | - Johanna Berg
- Department
of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Jens Kurreck
- Department
of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Ahmed S. G. Khalil
- Environmental
and Smart Technology Group (ESTG), Faculty of Science, Fayoum University, 63514 Fayoum, Egypt
- Materials
Science & Engineering Department, School of Innovative Design
Engineering, Egypt-Japan University of Science
and Technology (E-JUST), 21934 Alexandria, Egypt
| |
Collapse
|
31
|
Xiang Y, Miller K, Guan J, Kiratitanaporn W, Tang M, Chen S. 3D bioprinting of complex tissues in vitro: state-of-the-art and future perspectives. Arch Toxicol 2022; 96:691-710. [PMID: 35006284 PMCID: PMC8850226 DOI: 10.1007/s00204-021-03212-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
The pharmacology and toxicology of a broad variety of therapies and chemicals have significantly improved with the aid of the increasing in vitro models of complex human tissues. Offering versatile and precise control over the cell population, extracellular matrix (ECM) deposition, dynamic microenvironment, and sophisticated microarchitecture, which is desired for the in vitro modeling of complex tissues, 3D bio-printing is a rapidly growing technology to be employed in the field. In this review, we will discuss the recent advancement of printing techniques and bio-ink sources, which have been spurred on by the increasing demand for modeling tactics and have facilitated the development of the refined tissue models as well as the modeling strategies, followed by a state-of-the-art update on the specialized work on cancer, heart, muscle and liver. In the end, the toxicological modeling strategies, substantial challenges, and future perspectives for 3D printed tissue models were explored.
Collapse
Affiliation(s)
- Yi Xiang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Kathleen Miller
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA
| | | | - Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, USA.
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA.
| |
Collapse
|
32
|
Vrana NE, Gupta S, Mitra K, Rizvanov AA, Solovyeva VV, Antmen E, Salehi M, Ehterami A, Pourchet L, Barthes J, Marquette CA, von Unge M, Wang CY, Lai PL, Bit A. From 3D printing to 3D bioprinting: the material properties of polymeric material and its derived bioink for achieving tissue specific architectures. Cell Tissue Bank 2022; 23:417-440. [PMID: 35000046 DOI: 10.1007/s10561-021-09975-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/31/2021] [Indexed: 12/22/2022]
Abstract
The application of 3D printing technologies fields for biological tissues, organs, and cells in the context of medical and biotechnology applications requires a significant amount of innovation in a narrow printability range. 3D bioprinting is one such way of addressing critical design challenges in tissue engineering. In a more general sense, 3D printing has become essential in customized implant designing, faithful reproduction of microenvironmental niches, sustainable development of implants, in the capacity to address issues of effective cellular integration, and long-term stability of the cellular constructs in tissue engineering. This review covers various aspects of 3D bioprinting, describes the current state-of-the-art solutions for all aforementioned critical issues, and includes various illustrative representations of technologies supporting the development of phases of 3D bioprinting. It also demonstrates several bio-inks and their properties crucial for being used for 3D printing applications. The review focus on bringing together different examples and current trends in tissue engineering applications, including bone, cartilage, muscles, neuron, skin, esophagus, trachea, tympanic membrane, cornea, blood vessel, immune system, and tumor models utilizing 3D printing technology and to provide an outlook of the future potentials and barriers.
Collapse
Affiliation(s)
| | | | - Kunal Mitra
- Florida Institute of Technology, Melbourne, USA
| | | | | | - Ezgi Antmen
- Center of Excellence in Biomaterials and Tissue Engineering, BIOMATEN, Middle East Technical University (METU), Ankara, Turkey
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Arian Ehterami
- Department of Mechanical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Lea Pourchet
- UMR 1121, Biomaterials and Bioengineering, INSERM, Strasbourg, France
| | - Julien Barthes
- UMR 1121, Biomaterials and Bioengineering, INSERM, Strasbourg, France
| | | | - Magnus von Unge
- Akershus University Hospital and University of Oslo, Oslo, Norway.,Center for Clinical Research, Uppsala University, Vasteras, Uppsala, Sweden
| | - Chi-Yun Wang
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Po-Liang Lai
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Arindam Bit
- National Institute of Technology, Raipur, India.
| |
Collapse
|
33
|
Engineering Biological Tissues from the Bottom-Up: Recent Advances and Future Prospects. MICROMACHINES 2021; 13:mi13010075. [PMID: 35056239 PMCID: PMC8780533 DOI: 10.3390/mi13010075] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023]
Abstract
Tissue engineering provides a powerful solution for current organ shortages, and researchers have cultured blood vessels, heart tissues, and bone tissues in vitro. However, traditional top-down tissue engineering has suffered two challenges: vascularization and reconfigurability of functional units. With the continuous development of micro-nano technology and biomaterial technology, bottom-up tissue engineering as a promising approach for organ and tissue modular reconstruction has gradually developed. In this article, relevant advances in living blocks fabrication and assembly techniques for creation of higher-order bioarchitectures are described. After a critical overview of this technology, a discussion of practical challenges is provided, and future development prospects are proposed.
Collapse
|
34
|
Dai M, Belaïdi JP, Fleury G, Garanger E, Rielland M, Schultze X, Lecommandoux S. Elastin-like Polypeptide-Based Bioink: A Promising Alternative for 3D Bioprinting. Biomacromolecules 2021; 22:4956-4966. [PMID: 34751573 DOI: 10.1021/acs.biomac.1c00861] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Three-dimensional (3D) bioprinting offers a great alternative to traditional techniques in tissue reconstruction, based on seeding cells manually into a scaffold, to better reproduce organs' complexity. When a suitable bioink is engineered with appropriate physicochemical properties, such a process can advantageously provide a spatial control of the patterning that improves tissue reconstruction. The design of an adequate bioink must fulfill a long list of criteria including biocompatibility, printability, and stability. In this context, we have developed a bioink containing a precisely controlled recombinant biopolymer, namely, elastin-like polypeptide (ELP). This material was further chemoselectively modified with cross-linkable moieties to provide a 3D network through photopolymerization. ELP chains were additionally either functionalized with a peptide sequence Gly-Arg-Gly-Asp-Ser (GRGDS) or combined with collagen I to enable cell adhesion. Our ELP-based bioinks were found to be printable, while providing excellent mechanical properties such as stiffness and elasticity in their cross-linked form. Besides, they were demonstrated to be biocompatible, showing viability and adhesion of dermal normal human fibroblasts (NHF). Expressions of specific extracellular matrix (ECM) protein markers as pro-collagen I, elastin, fibrillin, and fibronectin were revealed within the 3D network containing cells after only 18 days of culture, showing the great potential of ELP-based bioinks for tissue engineering.
Collapse
Affiliation(s)
- Michèle Dai
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France.,Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Jean-Philippe Belaïdi
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - Guillaume Fleury
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Elisabeth Garanger
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Maïté Rielland
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - Xavier Schultze
- L'Oréal Recherche Avancée, 1 avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | | |
Collapse
|
35
|
Shanti A, Hallfors N, Petroianu GA, Planelles L, Stefanini C. Lymph Nodes-On-Chip: Promising Immune Platforms for Pharmacological and Toxicological Applications. Front Pharmacol 2021; 12:711307. [PMID: 34483920 PMCID: PMC8415712 DOI: 10.3389/fphar.2021.711307] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Organs-on-chip are gaining increasing attention as promising platforms for drug screening and testing applications. However, lymph nodes-on-chip options remain limited although the lymph node is one of the main determinants of the immunotoxicity of newly developed pharmacological drugs. In this review, we describe existing biomimetic lymph nodes-on-chip, their design, and their physiological relevance to pharmacology and shed the light on future directions associated with lymph node-on-chip design and implementation in drug discovery and development.
Collapse
Affiliation(s)
- Aya Shanti
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nicholas Hallfors
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Georg A Petroianu
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Lourdes Planelles
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
36
|
Luttrell SM, Smith AST, Mack DL. Creating stem cell-derived neuromuscular junctions in vitro. Muscle Nerve 2021; 64:388-403. [PMID: 34328673 PMCID: PMC9292444 DOI: 10.1002/mus.27360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Recent development of novel therapies has improved mobility and quality of life for people suffering from inheritable neuromuscular disorders. Despite this progress, the majority of neuromuscular disorders are still incurable, in part due to a lack of predictive models of neuromuscular junction (NMJ) breakdown. Improvement of predictive models of a human NMJ would be transformative in terms of expanding our understanding of the mechanisms that underpin development, maintenance, and disease, and as a testbed with which to evaluate novel therapeutics. Induced pluripotent stem cells (iPSCs) are emerging as a clinically relevant and non‐invasive cell source to create human NMJs to study synaptic development and maturation, as well as disease modeling and drug discovery. This review will highlight the recent advances and remaining challenges to generating an NMJ capable of eliciting contraction of stem cell‐derived skeletal muscle in vitro. We explore the advantages and shortcomings of traditional NMJ culturing platforms, as well as the pioneering technologies and novel, biomimetic culturing systems currently in use to guide development and maturation of the neuromuscular synapse and extracellular microenvironment. Then, we will explore how this NMJ‐in‐a‐dish can be used to study normal assembly and function of the efferent portion of the neuromuscular arc, and how neuromuscular disease‐causing mutations disrupt structure, signaling, and function.
Collapse
Affiliation(s)
- Shawn M Luttrell
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Alec S T Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - David L Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
37
|
Kim D, Lee SJ, Youn J, Hong H, Eom S, Kim DS. A deep and permeable nanofibrous oval-shaped microwell array for the stable formation of viable and functional spheroids. Biofabrication 2021; 13. [PMID: 34030141 DOI: 10.1088/1758-5090/ac044c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/24/2021] [Indexed: 12/26/2022]
Abstract
Despite the potential of a nanofibrous (NF) microwell array as a permeable microwell array to improve the viability and functions of spheroids, thanks to the superior permeability to both gases and solutes, there have still been difficulties regarding the stable formation of spheroids in the NF microwell array due to the low aspect ratio (AR) and the large interspacing between microwells. This study proposes a nanofibrous oval-shaped microwell array, named the NOVA microwell array, with both a high AR and a high well density, enabling us to not only collect cells in the microwell with a high cell seeding efficiency, but also to generate multiple viable and functional spheroids in a uniform and stable manner. To realize a deep NOVA microwell array with a high aspect ratio (AR = 0.9) and a high well density (494 wells cm-2), we developed a matched-mold thermoforming process for the fabrication of both size- and AR-controllable NOVA microwell arrays with various interspacing between microwells while maintaining the porous nature of the NF membrane. The human hepatocellular carcinoma (HepG2) cell spheroids cultured on the deep NOVA microwell array not only had uniform size and shape, with a spheroid circularity of 0.80 ± 0.03 at a cell seeding efficiency of 94.29 ± 9.55%, but also exhibited enhanced viability with a small fraction of dead cells and promoted functionality with increased albumin secretion, compared with the conventional impermeable microwell array. The superior characteristics of the deep NOVA microwell array, i.e. a high AR, a high well density, and a high permeability, pave the way to the production of various viable and functional spheroids and even organoids in a scalable manner.
Collapse
Affiliation(s)
- Dohui Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seong Jin Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hyeonjun Hong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seongsu Eom
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
38
|
Abdel-Raouf KMA, Rezgui R, Stefanini C, Teo JCM, Christoforou N. Transdifferentiation of Human Fibroblasts into Skeletal Muscle Cells: Optimization and Assembly into Engineered Tissue Constructs through Biological Ligands. BIOLOGY 2021; 10:biology10060539. [PMID: 34208436 PMCID: PMC8235639 DOI: 10.3390/biology10060539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Engineered human skeletal muscle tissue is a platform tool that can help scientists and physicians better understand human physiology, pharmacology, and disease modeling. Over the past few years this area of research has been actively being pursued by many labs worldwide. Significant challenges remain, including accessing an adequate cell source, and achieving proper physiological-like architecture of the engineered tissue. To address cell resourcing we aimed at further optimizing a process called transdifferentiation which involves the direct conversion of fibroblasts into skeletal muscle cells. The opportunity here is that fibroblasts are readily available and can be expanded sufficiently to meet the needs of a tissue engineering approach. Additionally, we aimed to demonstrate the applicability of transdifferentiation in assembling tissue engineered skeletal muscle. We implemented a screening process of protein ligands in an effort to refine transdifferentiation, and identified that most proteins resulted in a deficit in transdifferentiation efficiency, although one resulted in robust expansion of cultured cells. We were also successful in assembling engineered constructs consisting of transdifferentiated cells. Future directives involve demonstrating that the engineered tissues are capable of contractile and functional activity, and pursuit of optimizing factors such as electrical and chemical exposure, towards achieving physiological parameters observed in human muscle. Abstract The development of robust skeletal muscle models has been challenging due to the partial recapitulation of human physiology and architecture. Reliable and innovative 3D skeletal muscle models recently described offer an alternative that more accurately captures the in vivo environment but require an abundant cell source. Direct reprogramming or transdifferentiation has been considered as an alternative. Recent reports have provided evidence for significant improvements in the efficiency of derivation of human skeletal myotubes from human fibroblasts. Herein we aimed at improving the transdifferentiation process of human fibroblasts (tHFs), in addition to the differentiation of murine skeletal myoblasts (C2C12), and the differentiation of primary human skeletal myoblasts (HSkM). Differentiating or transdifferentiating cells were exposed to single or combinations of biological ligands, including Follistatin, GDF8, FGF2, GDF11, GDF15, hGH, TMSB4X, BMP4, BMP7, IL6, and TNF-α. These were selected for their critical roles in myogenesis and regeneration. C2C12 and tHFs displayed significant differentiation deficits when exposed to FGF2, BMP4, BMP7, and TNF-α, while proliferation was significantly enhanced by FGF2. When exposed to combinations of ligands, we observed consistent deficit differentiation when TNF-α was included. Finally, our direct reprogramming technique allowed for the assembly of elongated, cross-striated, and aligned tHFs within tissue-engineered 3D skeletal muscle constructs. In conclusion, we describe an efficient system to transdifferentiate human fibroblasts into myogenic cells and a platform for the generation of tissue-engineered constructs. Future directions will involve the evaluation of the functional characteristics of these engineered tissues.
Collapse
Affiliation(s)
- Khaled M. A. Abdel-Raouf
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Department of Biology, American University in Cairo, New Cairo 11835, Egypt
- Correspondence: (K.M.A.A.-R.); (N.C.)
| | - Rachid Rezgui
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Cesare Stefanini
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Jeremy C. M. Teo
- Department of Mechanical and Biomedical Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Nicolas Christoforou
- Pfizer Inc., Rare Disease Research Unit, 610 Main Street, Cambridge, MA 02139, USA
- Correspondence: (K.M.A.A.-R.); (N.C.)
| |
Collapse
|
39
|
Varone A, Nguyen JK, Leng L, Barrile R, Sliz J, Lucchesi C, Wen N, Gravanis A, Hamilton GA, Karalis K, Hinojosa CD. A novel organ-chip system emulates three-dimensional architecture of the human epithelia and the mechanical forces acting on it. Biomaterials 2021; 275:120957. [PMID: 34130145 DOI: 10.1016/j.biomaterials.2021.120957] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 05/22/2021] [Accepted: 05/29/2021] [Indexed: 12/22/2022]
Abstract
Successful translation of in vivo experimental data to human patients is an unmet need and a bottleneck in the development of effective therapeutics. Organ-on-Chip technology aims to address this need by leveraging recent significant advancements in microfabrication and biomaterials, which enable modeling of organs and their functionality. These microengineered chips offer researchers the possibility to recreate critical elements of native tissue architecture such as in vivo relevant tissue-tissue interface, air-liquid interface, and mechanical forces, including mechanical stretch and fluidic shear stress, which are crucial to recapitulate tissue level functions. Here, we present the development of a new, comprehensive 3D cell-culture system, where we combined our proprietary Organ-Chip technology with the advantages offered by three-dimensional organotypic culture. Leveraging microfabrication techniques, we engineered a flexible chip that consists of a chamber containing an organotypic epithelium, surrounded by two vacuum channels that can be actuated to stretch the hydrogel throughout its thickness. Furthermore, the ceiling of this chamber is a removable lid with a built-in microchannel that can be perfused with liquid or air and removed as needed for direct access to the tissue. The bottom part of this chamber is made from a porous flexible membrane which allows diffusive mass transport to and from the microfluidic channel positioned below the membrane. This additional microfluidic channel can be coated with endothelial cells to emulate a blood vessel and recapitulate endothelial interactions. Our results show that the Open-Top Chip design successfully addresses common challenges associated with the Organs-on-Chip technology, including the capability to incorporate a tissue-specific extracellular matrix gel seeded with primary stromal cells, to reproduce the architectural complexity of tissues by micropatterning the gel, and to extract the gel for H&E staining. We also provide proof-of-concept data on the feasibility of using the system with primary human skin and alveolar epithelial cells.
Collapse
Affiliation(s)
- Antonio Varone
- Emulate Inc., 27 Drydock Avenue, 5th Floor, Boston, MA, 02210, USA; University of Crete Medical School, Department of Pharmacology, Heraklion, 71110, Greece.
| | - Justin Ke Nguyen
- Emulate Inc., 27 Drydock Avenue, 5th Floor, Boston, MA, 02210, USA
| | - Lian Leng
- Emulate Inc., 27 Drydock Avenue, 5th Floor, Boston, MA, 02210, USA
| | - Riccardo Barrile
- University of Cincinnati, Department of Biomedical Engineering, Cincinnati, OH, 45221, USA
| | - Josiah Sliz
- Emulate Inc., 27 Drydock Avenue, 5th Floor, Boston, MA, 02210, USA
| | | | - Norman Wen
- Emulate Inc., 27 Drydock Avenue, 5th Floor, Boston, MA, 02210, USA
| | - Achille Gravanis
- University of Crete Medical School, Department of Pharmacology, Heraklion, 71110, Greece
| | | | - Katia Karalis
- Emulate Inc., 27 Drydock Avenue, 5th Floor, Boston, MA, 02210, USA
| | | |
Collapse
|
40
|
Abdel-Tawab M. Considerations to Be Taken When Carrying Out Medicinal Plant Research-What We Learn from an Insight into the IC 50 Values, Bioavailability and Clinical Efficacy of Exemplary Anti-Inflammatory Herbal Components. Pharmaceuticals (Basel) 2021; 14:437. [PMID: 34066427 PMCID: PMC8148151 DOI: 10.3390/ph14050437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/23/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Medicinal plants represent a big reservoir for discovering new drugs against all kinds of diseases including inflammation. In spite the large number of promising anti-inflammatory plant extracts and isolated components, research on medicinal plants proves to be very difficult. Based on that background this review aims to provide a summarized insight into the hitherto known pharmacologically active concentrations, bioavailability, and clinical efficacy of boswellic acids, curcumin, quercetin and resveratrol. These examples have in common that the achieved plasma concentrations were found to be often far below the determined IC50 values in vitro. On the other hand demonstrated therapeutic effects suggest a necessity of rethinking our pharmacokinetic understanding. In this light this review discusses the value of plasma levels as pharmacokinetic surrogates in comparison to the more informative value of tissue concentrations. Furthermore the need for new methodological approaches is addressed like the application of combinatorial approaches for identifying and pharmacokinetic investigations of active multi-components. Also the physiological relevance of exemplary in vitro assays and absorption studies in cell-line based models is discussed. All these topics should be ideally considered to avoid inaccurate predictions for the efficacy of herbal components in vivo and to unlock the "black box" of herbal mixtures.
Collapse
Affiliation(s)
- Mona Abdel-Tawab
- Central Laboratory of German Pharmacists, Carl-Mannich-Str. 20, 65760 Eschborn, Germany; ; Tel.: +49-6196-937-955
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| |
Collapse
|
41
|
De Pieri A, Korman BD, Jüngel A, Wuertz-Kozak K. Engineering Advanced In Vitro Models of Systemic Sclerosis for Drug Discovery and Development. Adv Biol (Weinh) 2021; 5:e2000168. [PMID: 33852183 PMCID: PMC8717409 DOI: 10.1002/adbi.202000168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
| | - Benjamin D Korman
- Prof. B. D. Korman, Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Astrid Jüngel
- Prof. A. Jüngel, Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, 8008, Switzerland
- Prof. A. Jüngel, Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zurich, 8008, Switzerland
| | - Karin Wuertz-Kozak
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
- Prof. K. Wuertz-Kozak, Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, 81547, Germany
| |
Collapse
|
42
|
Engineered microtissues for the bystander therapy against cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111854. [PMID: 33579487 DOI: 10.1016/j.msec.2020.111854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
Thymidine kinase expressing human adipose mesenchymal stem cells (TK-hAMSCs) in combination with ganciclovir (GCV) are an effective platform for antitumor bystander therapy in mice models. However, this strategy requires multiple TK-hAMSCs administrations and a substantial number of cells. Therefore, for clinical translation, it is necessary to find a biocompatible scaffold providing TK-hAMSCs retention in the implantation site against their rapid wash-out. We have developed a microtissue (MT) composed by TKhAMSCs and a scaffold made of polylactic acid microparticles and cell-derived extracellular matrix deposited by hAMSCs. The efficacy of these MTs as vehicles for TK-hAMSCs/GCV bystander therapy was evaluated in a rodent model of human prostate cancer. Subcutaneously implanted MTs were integrated in the surrounding tissue, allowing neovascularization and maintenance of TK-hAMSCs viability. Furthermore, MTs implanted beside tumors allowed TK-hAMSCs migration towards tumor cells and, after GCV administration, inhibited tumor growth. These results indicate that TK-hAMSCs-MTs are promising cell reservoirs for clinical use of therapeutic MSCs in bystander therapies.
Collapse
|
43
|
Fritschen A, Blaeser A. Biosynthetic, biomimetic, and self-assembled vascularized Organ-on-a-Chip systems. Biomaterials 2020; 268:120556. [PMID: 33310539 DOI: 10.1016/j.biomaterials.2020.120556] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Organ-on-a-Chip (OOC) devices have seen major advances in the last years with respect to biological complexity, physiological composition and biomedical relevance. In this context, integration of vasculature has proven to be a crucial element for long-term culture of thick tissue samples as well as for realistic pharmacokinetic, toxicity and metabolic modelling. With the emergence of digital production technologies and the reinvention of existing tools, a multitude of design approaches for guided angio- and vasculogenesis is available today. The underlying production methods can be categorized into biosynthetic, biomimetic and self-assembled vasculature formation. The diversity and importance of production approaches, vascularization strategies as well as biomaterials and cell sourcing are illustrated in this work. A comprehensive technological review with a strong focus on the challenge of producing physiologically relevant vascular structures is given. Finally, the remaining obstacles and opportunities in the development of vascularized Organ-on-a-Chip platforms for advancing drug development and predictive disease modelling are noted.
Collapse
Affiliation(s)
- Anna Fritschen
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany.
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Germany.
| |
Collapse
|
44
|
Panzer KV, Burrell JC, Helm KVT, Purvis EM, Zhang Q, Le AD, O’Donnell JC, Cullen DK. Tissue Engineered Bands of Büngner for Accelerated Motor and Sensory Axonal Outgrowth. Front Bioeng Biotechnol 2020; 8:580654. [PMID: 33330416 PMCID: PMC7714719 DOI: 10.3389/fbioe.2020.580654] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Following peripheral nerve injury comprising a segmental defect, the extent of axon regeneration decreases precipitously with increasing gap length. Schwann cells play a key role in driving axon re-growth by forming aligned tubular guidance structures called bands of Büngner, which readily occurs in distal nerve segments as well as within autografts - currently the most reliable clinically-available bridging strategy. However, host Schwann cells generally fail to infiltrate large-gap acellular scaffolds, resulting in markedly inferior outcomes and motivating the development of next-generation bridging strategies capable of fully exploiting the inherent pro-regenerative capability of Schwann cells. We sought to create preformed, implantable Schwann cell-laden microtissue that emulates the anisotropic structure and function of naturally-occurring bands of Büngner. Accordingly, we developed a biofabrication scheme leveraging biomaterial-induced self-assembly of dissociated rat primary Schwann cells into dense, fiber-like three-dimensional bundles of Schwann cells and extracellular matrix within hydrogel micro-columns. This engineered microtissue was found to be biomimetic of morphological and phenotypic features of endogenous bands of Büngner, and also demonstrated 8 and 2× faster rates of axonal extension in vitro from primary rat spinal motor neurons and dorsal root ganglion sensory neurons, respectively, compared to 3D matrix-only controls or planar Schwann cells. To our knowledge, this is the first report of accelerated motor axon outgrowth using aligned Schwann cell constructs. For translational considerations, this microtissue was also fabricated using human gingiva-derived Schwann cells as an easily accessible autologous cell source. These results demonstrate the first tissue engineered bands of Büngner (TE-BoBs) comprised of dense three-dimensional bundles of longitudinally aligned Schwann cells that are readily scalable as implantable grafts to accelerate axon regeneration across long segmental nerve defects.
Collapse
Affiliation(s)
- Kate V. Panzer
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Justin C. Burrell
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Kaila V. T. Helm
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Erin M. Purvis
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral and Maxillofacial Surgery, Penn Medicine Hospital of University of Pennsylvania, Philadelphia, PA, United States
| | - Anh D. Le
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral and Maxillofacial Surgery, Penn Medicine Hospital of University of Pennsylvania, Philadelphia, PA, United States
| | - John C. O’Donnell
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
45
|
Kim J, Adachi T. Modulation of Sost Gene Expression Under Hypoxia in Three-Dimensional Scaffold-Free Osteocytic Tissue. Tissue Eng Part A 2020; 27:1037-1043. [PMID: 33040693 DOI: 10.1089/ten.tea.2020.0228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Bone-related studies have been widely carried out by culturing cells on two-dimensional (2D) culture system because of its easiness of handling, but these 2D in vitro achievements may imply a distinct outcome compared with the in vivo situation. On the contrary, three-dimensional (3D) culture system has been suggested as a better biomimetic in vitro model by providing an appropriate cell-cell or cell-matrix interaction. In this study, we successfully reconstructed a 3D disk type of scaffold-free tissue (SFT) using mouse osteoblast-like cells, which evoked an osteocyte differentiation within 2 days. Particularly, the SFT was also utilized as an in vitro osteocytic model to elucidate the effect of hypoxia on cellular differentiation capability. As a result, the hypoxia upregulated a matured osteocyte marker, Sost, in the SFT, whereas both osteoblast and osteocyte markers were significantly downregulated by hypoxia in the 2D conventional monolayer model. The results imply that the hypoxia may enhance the initiation of osteocyte differentiation and retain the osteocyte differentiation in the 3D culture system. Of note, we reported the significance of 3D culture system that might represent the in vivo situation regarding cellular response to stimuli. Hence, our study suggests wide applications of SFT using osteoblast cells as a novel in vitro osteocyte model for the osteocyte-related studies. Impact statement In this study, we fabricated a three-dimensional (3D) disk type of scaffold-free osteocytic tissue, termed scaffold-free tissue (SFT), reconstructed by mouse osteoblast-like cells. It induced an osteocyte differentiation of osteoblast-like cells in the SFT within 2 days. Moreover, we first showed that a matured osteocyte marker, Sost, was modulated by hypoxia in the SFT in a different manner compared with the two-dimensional (2D) monolayer. These results highlighted the significance of 3D culture system that might represent the in vivo situation regarding cellular response to stimuli. Of note, our model can be utilized as a new in vitro osteocyte model for the osteocyte-related studies.
Collapse
Affiliation(s)
- Jeonghyun Kim
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Taiji Adachi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
Xie Z, Gao M, Lobo AO, Webster TJ. 3D Bioprinting in Tissue Engineering for Medical Applications: The Classic and the Hybrid. Polymers (Basel) 2020; 12:E1717. [PMID: 32751797 PMCID: PMC7464247 DOI: 10.3390/polym12081717] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Three-dimensional (3D) printing, as one of the most popular recent additive manufacturing processes, has shown strong potential for the fabrication of biostructures in the field of tissue engineering, most notably for bones, orthopedic tissues, and associated organs. Desirable biological, structural, and mechanical properties can be achieved for 3D-printed constructs with a proper selection of biomaterials and compatible bioprinting methods, possibly even while combining additive and conventional manufacturing (AM and CM) procedures. However, challenges remain in the need for improved printing resolution (especially at the nanometer level), speed, and biomaterial compatibilities, and a broader range of suitable 3D-printed materials. This review provides an overview of recent advances in the development of 3D bioprinting techniques, particularly new hybrid 3D bioprinting technologies for combining the strengths of both AM and CM, along with a comprehensive set of material selection principles, promising medical applications, and limitations and future prospects.
Collapse
Affiliation(s)
- Zelong Xie
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; (Z.X.); (M.G.)
| | - Ming Gao
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; (Z.X.); (M.G.)
| | - Anderson O. Lobo
- LIMAV–Interdisciplinary Laboratory for Advanced Materials, BioMatLab, UFPI–Federal University of Piauí, Teresina 64049-550, Brazil;
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; (Z.X.); (M.G.)
| |
Collapse
|
47
|
Calar K, Plesselova S, Bhattacharya S, Jorgensen M, de la Puente P. Human Plasma-Derived 3D Cultures Model Breast Cancer Treatment Responses and Predict Clinically Effective Drug Treatment Concentrations. Cancers (Basel) 2020; 12:cancers12071722. [PMID: 32610529 PMCID: PMC7407241 DOI: 10.3390/cancers12071722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023] Open
Abstract
Lack of efficacy and a low overall success rate of phase I-II clinical trials are the most common failures when it comes to advancing cancer treatment. Current drug sensitivity screenings present several challenges including differences in cell growth rates, the inconsistent use of drug metrics, and the lack of translatability. Here, we present a patient-derived 3D culture model to overcome these limitations in breast cancer (BCa). The human plasma-derived 3D culture model (HuP3D) utilizes patient plasma as the matrix, where BCa cell lines and primary BCa biopsies were grown and screened for drug treatments. Several drug metrics were evaluated from relative cell count and growth rate curves. Correlations between HuP3D metrics, established preclinical models, and clinical effective concentrations in patients were determined. HuP3D efficiently supported the growth and expansion of BCa cell lines and primary breast cancer tumors as both organoids and single cells. Significant and strong correlations between clinical effective concentrations in patients were found for eight out of ten metrics for HuP3D, while a very poor positive correlation and a moderate correlation was found for 2D models and other 3D models, respectively. HuP3D is a feasible and efficacious platform for supporting the growth and expansion of BCa, allowing high-throughput drug screening and predicting clinically effective therapies better than current preclinical models.
Collapse
Affiliation(s)
- Kristin Calar
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
| | - Simona Plesselova
- Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain;
| | - Somshuvra Bhattacharya
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
| | - Megan Jorgensen
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
- MD/PhD Program, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA
| | - Pilar de la Puente
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
- Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA
- Flow Cytometry Core, Sanford Research, Sioux Falls, SD 57104, USA
- Correspondence: ; Tel.: +1-605-312-6042
| |
Collapse
|
48
|
Pakhomova C, Popov D, Maltsev E, Akhatov I, Pasko A. Software for Bioprinting. Int J Bioprint 2020; 6:279. [PMID: 33088988 PMCID: PMC7557344 DOI: 10.18063/ijb.v6i3.279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
The bioprinting of heterogeneous organs is a crucial issue. To reach the complexity of such organs, there is a need for highly specialized software that will meet all requirements such as accuracy, complexity, and others. The primary objective of this review is to consider various software tools that are used in bioprinting and to reveal their capabilities. The sub-objective was to consider different approaches for the model creation using these software tools. Related articles on this topic were analyzed. Software tools are classified based on control tools, general computer-aided design (CAD) tools, tools to convert medical data to CAD formats, and a few highly specialized research-project tools. Different geometry representations are considered, and their advantages and disadvantages are considered applicable to heterogeneous volume modeling and bioprinting. The primary factor for the analysis is suitability of the software for heterogeneous volume modeling and bioprinting or multimaterial three-dimensional printing due to the commonality of these technologies. A shortage of specialized suitable software tools is revealed. There is a need to develop a new application area such as computer science for bioprinting which can contribute significantly in future research work.
Collapse
Affiliation(s)
- Catherine Pakhomova
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow.,Institute of Engineering Physics for Biomedicine, NRNU Mephi, Moscow
| | - Dmitry Popov
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow
| | - Eugenii Maltsev
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow
| | - Iskander Akhatov
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow
| | - Alexander Pasko
- Center for Design, Manufacturing and Materials, Skolkovo Institute of Science and Technology, Moscow.,The National Centre for Computer Animation, Bournemouth University, UK
| |
Collapse
|
49
|
Pitoulis FG, Watson SA, Perbellini F, Terracciano CM. Myocardial slices come to age: an intermediate complexity in vitro cardiac model for translational research. Cardiovasc Res 2020; 116:1275-1287. [PMID: 31868875 PMCID: PMC7243278 DOI: 10.1093/cvr/cvz341] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/31/2019] [Accepted: 12/19/2019] [Indexed: 12/17/2022] Open
Abstract
Although past decades have witnessed significant reductions in mortality of heart failure together with advances in our understanding of its cellular, molecular, and whole-heart features, a lot of basic cardiac research still fails to translate into clinical practice. In this review we examine myocardial slices, a novel model in the translational arena. Myocardial slices are living ultra-thin sections of heart tissue. Slices maintain the myocardium's native function (contractility, electrophysiology) and structure (multicellularity, extracellular matrix) and can be prepared from animal and human tissue. The discussion begins with the history and current advances in the model, the different interlaboratory methods of preparation and their potential impact on results. We then contextualize slices' advantages and limitations by comparing it with other cardiac models. Recently, sophisticated methods have enabled slices to be cultured chronically in vitro while preserving the functional and structural phenotype. This is more timely now than ever where chronic physiologically relevant in vitro platforms for assessment of therapeutic strategies are urgently needed. We interrogate the technological developments that have permitted this, their limitations, and future directions. Finally, we look into the general obstacles faced by the translational field, and how implementation of research systems utilizing slices could help in resolving these.
Collapse
Affiliation(s)
- Fotios G Pitoulis
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Samuel A Watson
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Filippo Perbellini
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cesare M Terracciano
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
50
|
Xie M, Gao Q, Fu J, Chen Z, He Y. Bioprinting of novel 3D tumor array chip for drug screening. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00078-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|