1
|
Tusnim J, Kutuzov P, Grasman JM. In Vitro Models for Peripheral Nerve Regeneration. Adv Healthc Mater 2024:e2401605. [PMID: 39324286 DOI: 10.1002/adhm.202401605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Peripheral nerve injury (PNI) resulting in lesions is highly prevalent clinically, but current therapeutic approaches fail to provide satisfactory outcomes in many patients. While peripheral nerves have intrinsic regenerative capacity, the regenerative capabilities of peripheral nerves are often insufficient to restore full functionality. This highlights an unmet need for developing more effective strategies to repair damaged peripheral nerves and improve regenerative success. Consequently, researchers are actively exploring a variety of therapeutic strategies, encompassing the local delivery of trophic factors or bioactive molecules, the design of advanced biomaterials that interact with regenerating axons, and augmentation with nerve guidance conduits or complex prostheses. However, clinical translation of these technologies remains limited, emphasizing the need for continued research on peripheral nerve regeneration modalities that can enhance functional restoration. Experimental models that accurately recapitulate key aspects of peripheral nerve injury and repair biology can accelerate therapeutic development by enabling systematic testing of new techniques. Advancing regenerative therapies for PNI requires bridging the gap between basic science discoveries and clinical application. This review discusses different in vitro models of peripheral nerve injury and repair, including their advantages, limitations, and potential applications.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Peter Kutuzov
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
2
|
Modi PS, Singh A, Chaturvedi A, Agarwal S, Dutta R, Nayak R, Singh AK. Tissue chips as headway model and incitement technology. Synth Syst Biotechnol 2024; 10:86-101. [PMID: 39286054 PMCID: PMC11403008 DOI: 10.1016/j.synbio.2024.08.007] [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: 01/12/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Tissue on a chip or organ-on-chip (OOC) is a technology that's dignified to form a transformation in drug discovery through the use of advanced platforms. These are 3D in-vitro cell culture models that mimic micro-environment of human organs or tissues on artificial microstructures built on a portable microfluidic chip without involving sacrificial humans or animals. This review article aims to offer readers a thorough and insightful understanding of technology. It begins with an in-depth understanding of chip design and instrumentation, underlining its pivotal role and the imperative need for its development in the modern scientific landscape. The review article explores into the myriad applications of OOC technology, showcasing its transformative impact on fields such as radiobiology, drug discovery and screening, and its pioneering use in space research. In addition to highlighting these diverse applications, the article provides a critical analysis of the current challenges that OOC technology faces. It examines both the biological and technical limitations that hinder its progress and efficacy and discusses the potential advancements and innovations that could drive the OOC technology forward. Through this comprehensive review, readers will gain a deep appreciation of the significance, capabilities, and evolving landscape of OOC technology.
Collapse
Affiliation(s)
- Prerna Suchitan Modi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Abhishek Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Awyang Chaturvedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Shailly Agarwal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Raghav Dutta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Alok Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| |
Collapse
|
3
|
Abed H, Radha R, Anjum S, Paul V, AlSawaftah N, Pitt WG, Ashammakhi N, Husseini GA. Targeted Cancer Therapy-on-A-Chip. Adv Healthc Mater 2024:e2400833. [PMID: 39101627 DOI: 10.1002/adhm.202400833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/15/2024] [Indexed: 08/06/2024]
Abstract
Targeted cancer therapy (TCT) is gaining increased interest because it reduces the risks of adverse side effects by specifically treating tumor cells. TCT testing has traditionally been performed using two-dimensional (2D) cell culture and animal studies. Organ-on-a-chip (OoC) platforms have been developed to recapitulate cancer in vitro, as cancer-on-a-chip (CoC), and used for chemotherapeutics development and testing. This review explores the use of CoCs to both develop and test TCTs, with a focus on three main aspects, the use of CoCs to identify target biomarkers for TCT development, the use of CoCs to test free, un-encapsulated TCTs, and the use of CoCs to test encapsulated TCTs. Despite current challenges such as system scaling, and testing externally triggered TCTs, TCToC shows a promising future to serve as a supportive, pre-clinical platform to expedite TCT development and bench-to-bedside translation.
Collapse
Affiliation(s)
- Heba Abed
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Remya Radha
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Shabana Anjum
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Vinod Paul
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - Nour AlSawaftah
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, MI, 48824, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - Ghaleb A Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| |
Collapse
|
4
|
Barker S, Dagys L, Levitt MH, Utz M. Efficient Parahydrogen-Induced 13C Hyperpolarization on a Microfluidic Device. J Am Chem Soc 2024; 146:18379-18386. [PMID: 38916928 PMCID: PMC11240250 DOI: 10.1021/jacs.4c03271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/26/2024]
Abstract
We show the direct production and detection of 13C-hyperpolarized fumarate by parahydrogen-induced polarization (PHIP) in a microfluidic lab-on-a-chip (LoC) device and achieve 8.5% 13C polarization. This is the first demonstration of 13C-hyperpolarization of a metabolite by PHIP in a microfluidic device. LoC technology allows the culture of mammalian cells in a highly controlled environment, providing an important tool for the life sciences. In-situ preparation of hyperpolarized metabolites greatly enhances the ability to quantify metabolic processes in such systems by microfluidic NMR. PHIP of 1H nuclei has been successfully implemented in microfluidic systems, with mass sensitivities in the range of pmol/s. However, metabolic NMR requires high-yield production of hyperpolarized metabolites with longer spin life times than is possible with 1H. This can be achieved by transfer of the polarization onto 13C nuclei, which exhibit much longer T1 relaxation times. We report an improved microfluidic PHIP device, optimized using a finite element model, that enables the direct and efficient production of 13C-hyperpolarized fumarate.
Collapse
Affiliation(s)
- Sylwia
J. Barker
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute
of Microstructure Technology, Karlsruhe
Institute of Technology, Karlsruhe 76131, Germany
| | - Laurynas Dagys
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute
of Chemical Physics, Vilnius University, Vilnius 01513, Lithuania
| | - Malcolm H. Levitt
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Marcel Utz
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute
of Microstructure Technology, Karlsruhe
Institute of Technology, Karlsruhe 76131, Germany
| |
Collapse
|
5
|
Mihaylova A, Shopova D, Parahuleva N, Yaneva A, Bakova D. (3D) Bioprinting-Next Dimension of the Pharmaceutical Sector. Pharmaceuticals (Basel) 2024; 17:797. [PMID: 38931464 PMCID: PMC11206453 DOI: 10.3390/ph17060797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/26/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
To create a review of the published scientific literature on the benefits and potential perspectives of the use of 3D bio-nitrification in the field of pharmaceutics. This work was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for reporting meta-analyses and systematic reviews. The scientific databases PubMed, Scopus, Google Scholar, and ScienceDirect were used to search and extract data using the following keywords: 3D bioprinting, drug research and development, personalized medicine, pharmaceutical companies, clinical trials, drug testing. The data points to several aspects of the application of bioprinting in pharmaceutics were reviewed. The main applications of bioprinting are in the development of new drug molecules as well as in the preparation of personalized drugs, but the greatest benefits are in terms of drug screening and testing. Growth in the field of 3D printing has facilitated pharmaceutical applications, enabling the development of personalized drug screening and drug delivery systems for individual patients. Bioprinting presents the opportunity to print drugs on demand according to the individual needs of the patient, making the shape, structure, and dosage suitable for each of the patient's physical conditions, i.e., print specific drugs for controlled release rates; print porous tablets to reduce swallowing difficulties; make transdermal microneedle patches to reduce patient pain; and so on. On the other hand, bioprinting can precisely control the distribution of cells and biomaterials to build organoids, or an Organ-on-a-Chip, for the testing of drugs on printed organs mimicking specified disease characteristics instead of animal testing and clinical trials. The development of bioprinting has the potential to offer customized drug screening platforms and drug delivery systems meeting a range of individualized needs, as well as prospects at different stages of drug development and patient therapy. The role of bioprinting in preclinical and clinical testing of drugs is also of significant importance in terms of shortening the time to launch a medicinal product on the market.
Collapse
Affiliation(s)
- Anna Mihaylova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Dobromira Shopova
- Department of Prosthetic Dentistry, Faculty of Dental Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Nikoleta Parahuleva
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Antoniya Yaneva
- Department of Medical Informatics, Biostatistics and eLearning, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Desislava Bakova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| |
Collapse
|
6
|
Safarzadeh M, Richardson LS, Kammala AK, Mosebarger A, Bettayeb M, Kim S, Lam PY, Radnaa E, Han A, Menon R. A multi-organ, feto-maternal interface organ-on-chip, models pregnancy pathology and is a useful preclinical extracellular vesicle drug trial platform. EXTRACELLULAR VESICLE 2024; 3:100035. [PMID: 38872854 PMCID: PMC11175617 DOI: 10.1016/j.vesic.2024.100035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Pregnant women and their fetuses are often excluded from clinical trials due to missing drug-related pre-clinical trial information at the human feto-maternal interface (FMi). The two interfaces-placenta/decidua and fetal membranes/decidua are gatekeepers of drug transport; however, testing their functions is impractical during pregnancy. Limitations of current in-vivo/in-vitro models have hampered drug development and testing during pregnancy. Hence, major complications like preterm births and maternal and neonatal mortalities remain high. Advancements in organ-on-chip (OOC) platforms to test drug kinetics and efficacy and novel extracellular vesicle-based fetal drug delivery are expected to accelerate preclinical trials related to pregnancy complications. Here we report the development and testing of a humanized multi-organ fetal membrane/placenta (fetal)-decidua (maternal) interface OOC (FMi-PLA-OOC) that contains seven cell types interconnected through microchannels to maintain intercellular interactions as seen in-utero. Cytotoxicity, propagation, mechanism of action, and efficacy of engineered extracellular vesicles containing anti-inflammatory interleukin (IL)-10 (eIL-10) were evaluated to reduce FMi inflammation associated with preterm birth. A healthy and disease model (lipopolysaccharide-infectious inflammation) of the FMi-PLA-OOC was created and co-treated with eIL-10. eIL-10 propagated from the maternal to fetal side within 72-hours, localized in all cell types, showed no cytotoxicity, activated IL-10 signaling pathways, and reduced lipopolysaccharide-induced inflammation (minimized NF-kB activation and proinflammatory cytokine production). These data recapitulated eIL-10s' ability to reduce inflammation and delay infection-associated preterm birth in mouse models, suggesting FMi-PLA-OOC as an alternative approach to using animal models. Additionally, we report the utility of eIL-10 that can traverse through FMis to reduce inflammation-associated pregnancy complications.
Collapse
Affiliation(s)
- Melody Safarzadeh
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Lauren S. Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Angela Mosebarger
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Mohamed Bettayeb
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Sungjin Kim
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Po Yi Lam
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Enkhtuya Radnaa
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Arum Han
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
7
|
Ravi K, Manoharan TJM, Wang KC, Pockaj B, Nikkhah M. Engineered 3D ex vivo models to recapitulate the complex stromal and immune interactions within the tumor microenvironment. Biomaterials 2024; 305:122428. [PMID: 38147743 PMCID: PMC11098715 DOI: 10.1016/j.biomaterials.2023.122428] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/28/2023]
Abstract
Cancer thrives in a complex environment where interactions between cellular and acellular components, surrounding the tumor, play a crucial role in disease development and progression. Despite significant progress in cancer research, the mechanism driving tumor growth and therapeutic outcomes remains elusive. Two-dimensional (2D) cell culture assays and in vivo animal models are commonly used in cancer research and therapeutic testing. However, these models suffer from numerous shortcomings including lack of key features of the tumor microenvironment (TME) & cellular composition, cost, and ethical clearance. To that end, there is an increased interest in incorporating and elucidating the influence of TME on cancer progression. Advancements in 3D-engineered ex vivo models, leveraging biomaterials and microengineering technologies, have provided an unprecedented ability to reconstruct native-like bioengineered cancer models to study the heterotypic interactions of TME with a spatiotemporal organization. These bioengineered cancer models have shown excellent capabilities to bridge the gap between oversimplified 2D systems and animal models. In this review article, we primarily provide an overview of the immune and stromal cellular components of the TME and then discuss the latest state-of-the-art 3D-engineered ex vivo platforms aiming to recapitulate the complex TME features. The engineered TME model, discussed herein, are categorized into three main sections according to the cellular interactions within TME: (i) Tumor-Stromal interactions, (ii) Tumor-Immune interactions, and (iii) Complex TME interactions. Finally, we will conclude the article with a perspective on how these models can be instrumental for cancer translational studies and therapeutic testing.
Collapse
Affiliation(s)
- Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | | | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | | | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
8
|
R N, Aggarwal A, Sravani AB, Mallya P, Lewis S. Organ-On-A-Chip: An Emerging Research Platform. Organogenesis 2023; 19:2278236. [PMID: 37965897 PMCID: PMC10653779 DOI: 10.1080/15476278.2023.2278236] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
In drug development, conventional preclinical and clinical testing stages rely on cell cultures and animal experiments, but these methods may fall short of fully representing human biology. To overcome this limitation, the emergence of organ-on-a-chip (OOC) technology has sparked interest as a transformative approach in drug testing research. By closely replicating human organ responses to external signals, OOC devices hold immense potential in revolutionizing drug efficacy and safety predictions. This review focuses on the advancements, applications, and prospects of OOC devices in drug testing. Based on the latest advances in the field of OOC systems and their clinical applications, this review reflects the effectiveness of OOC devices in replacing human volunteers in certain clinical studies. This review underscores the critical role of OOC technology in transforming drug testing methodologies.
Collapse
Affiliation(s)
- Nithin R
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Ayushi Aggarwal
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Anne Boyina Sravani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Pooja Mallya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
9
|
Carels N, Sgariglia D, Junior MGV, Lima CR, Carneiro FRG, da Silva GF, da Silva FAB, Scardini R, Tuszynski JA, de Andrade CV, Monteiro AC, Martins MG, da Silva TG, Ferraz H, Finotelli PV, Balbino TA, Pinto JC. A Strategy Utilizing Protein-Protein Interaction Hubs for the Treatment of Cancer Diseases. Int J Mol Sci 2023; 24:16098. [PMID: 38003288 PMCID: PMC10671768 DOI: 10.3390/ijms242216098] [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: 07/23/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 11/26/2023] Open
Abstract
We describe a strategy for the development of a rational approach of neoplastic disease therapy based on the demonstration that scale-free networks are susceptible to specific attacks directed against its connective hubs. This strategy involves the (i) selection of up-regulated hubs of connectivity in the tumors interactome, (ii) drug repurposing of these hubs, (iii) RNA silencing of non-druggable hubs, (iv) in vitro hub validation, (v) tumor-on-a-chip, (vi) in vivo validation, and (vii) clinical trial. Hubs are protein targets that are assessed as targets for rational therapy of cancer in the context of personalized oncology. We confirmed the existence of a negative correlation between malignant cell aggressivity and the target number needed for specific drugs or RNA interference (RNAi) to maximize the benefit to the patient's overall survival. Interestingly, we found that some additional proteins not generally targeted by drug treatments might justify the addition of inhibitors designed against them in order to improve therapeutic outcomes. However, many proteins are not druggable, or the available pharmacopeia for these targets is limited, which justifies a therapy based on encapsulated RNAi.
Collapse
Affiliation(s)
- Nicolas Carels
- Platform of Biological System Modeling, Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (C.R.L.); (G.F.d.S.)
| | - Domenico Sgariglia
- Engenharia de Sistemas e Computação, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-972, RJ, Brazil;
| | - Marcos Guilherme Vieira Junior
- Computational Modeling of Biological Systems, Scientific Computing Program (PROCC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil or (M.G.V.J.); (F.A.B.d.S.)
| | - Carlyle Ribeiro Lima
- Platform of Biological System Modeling, Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (C.R.L.); (G.F.d.S.)
| | - Flávia Raquel Gonçalves Carneiro
- Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (F.R.G.C.); (R.S.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, RJ, Brazil
| | - Gilberto Ferreira da Silva
- Platform of Biological System Modeling, Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (C.R.L.); (G.F.d.S.)
| | - Fabricio Alves Barbosa da Silva
- Computational Modeling of Biological Systems, Scientific Computing Program (PROCC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil or (M.G.V.J.); (F.A.B.d.S.)
| | - Rafaela Scardini
- Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (F.R.G.C.); (R.S.)
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, RJ, Brazil
- Centro de Ciências Biológicas e da Saúde (CCBS), Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro 22290-255, RJ, Brazil
| | - Jack Adam Tuszynski
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, 10129 Turin, Italy;
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
- Department of Physics, University of Alberta, Edmonton, AB T6G 2J1, Canada
| | - Cecilia Vianna de Andrade
- Department of Pathology, Instituto Fernandes Figueira, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 22250-020, RJ, Brazil;
| | - Ana Carolina Monteiro
- Laboratory of Osteo and Tumor Immunology, Department of Immunobiology, Fluminense Federal University, Rio de Janeiro 24210-201, RJ, Brazil;
| | - Marcel Guimarães Martins
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| | - Talita Goulart da Silva
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| | - Helen Ferraz
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| | - Priscilla Vanessa Finotelli
- Laboratório de Nanotecnologia Biofuncional, Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil;
| | - Tiago Albertini Balbino
- Nanotechnology Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil;
| | - José Carlos Pinto
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| |
Collapse
|
10
|
Lu Z, Miao X, Song Q, Ding H, Rajan SAP, Skardal A, Votanopoulos KI, Dai K, Zhao W, Lu B, Atala A. Detection of lineage-reprogramming efficiency of tumor cells in a 3D-printed liver-on-a-chip model. Theranostics 2023; 13:4905-4918. [PMID: 37771785 PMCID: PMC10526656 DOI: 10.7150/thno.86921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/04/2023] [Indexed: 09/30/2023] Open
Abstract
Background: The liver metastasis accompanied with the loss of liver function is one of the most common complications in patients with triple-negative breast cancers (TNBC). Lineage reprogramming, as a technique direct inducing the functional cell types from one lineage to another lineage without passing through an intermediate pluripotent stage, is promising in changing cell fates and overcoming the limitations of primary cells. However, most reprogramming techniques are derived from human fibroblasts, and whether cancer cells can be reversed into hepatocytes remains elusive. Methods: Herein, we simplify preparation of reprogramming reagents by expressing six transcriptional factors (HNF4A, FOXA2, FOXA3, ATF5, PROX1, and HNF1) from two lentiviral vectors, each expressing three factors. Then the virus was transduced into MDA-MB-231 cells to generated human induced hepatocyte-like cells (hiHeps) and single-cell sequencing was used to analyze the fate for the cells after reprogramming. Furthermore, we constructed a Liver-on-a-chip (LOC) model by bioprinting the Gelatin Methacryloyl hydrogel loaded with hepatocyte extracellular vesicles (GelMA-EV) bioink onto the microfluidic chip to assess the metastasis behavior of the reprogrammed TNBC cells under the 3D liver microenvironment in vitro. Results: The combination of the genes HNF4A, FOXA2, FOXA3, ATF5, PROX1 and HNF1A could reprogram MDA-MB-231 tumor cells into human-induced hepatocytes (hiHeps), limiting metastasis of these cells. Single-cell sequencing analysis showed that the oncogenes were significantly inhibited while the liver-specific genes were activated after lineage reprogramming. Finally, the constructed LOC model showed that the hepatic phenotypes of the reprogrammed cells could be observed, and the metastasis of embedded cancer cells could be inhibited under the liver microenvironment. Conclusion: Our findings demonstrate that reprogramming could be a promising method to produce hepatocytes and treat TNBC liver metastasis. And the LOC model could intimate the 3D liver microenvironment and assess the behavior of the reprogrammed TNBC cells.
Collapse
Affiliation(s)
- Zuyan Lu
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Xiangwan Miao
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Huifen Ding
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Shiny Amala Priya Rajan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Kerong Dai
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| |
Collapse
|
11
|
Deng S, Li C, Cao J, Cui Z, Du J, Fu Z, Yang H, Chen P. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics 2023; 13:4526-4558. [PMID: 37649608 PMCID: PMC10465229 DOI: 10.7150/thno.87266] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
Drug evaluation has always been an important area of research in the pharmaceutical industry. However, animal welfare protection and other shortcomings of traditional drug development models pose obstacles and challenges to drug evaluation. Organ-on-a-chip (OoC) technology, which simulates human organs on a chip of the physiological environment and functionality, and with high fidelity reproduction organ-level of physiology or pathophysiology, exhibits great promise for innovating the drug development pipeline. Meanwhile, the advancement in artificial intelligence (AI) provides more improvements for the design and data processing of OoCs. Here, we review the current progress that has been made to generate OoC platforms, and how human single and multi-OoCs have been used in applications, including drug testing, disease modeling, and personalized medicine. Moreover, we discuss issues facing the field, such as large data processing and reproducibility, and point to the integration of OoCs and AI in data analysis and automation, which is of great benefit in future drug evaluation. Finally, we look forward to the opportunities and challenges faced by the coupling of OoCs and AI. In summary, advancements in OoCs development, and future combinations with AI, will eventually break the current state of drug evaluation.
Collapse
Affiliation(s)
- Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jiang Du
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
| | - Zheng Fu
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| |
Collapse
|
12
|
Huang YL, Dickerson LK, Kenerson H, Jiang X, Pillarisetty V, Tian Q, Hood L, Gujral TS, Yeung RS. Organotypic Models for Functional Drug Testing of Human Cancers. BME FRONTIERS 2023; 4:0022. [PMID: 37849667 PMCID: PMC10275620 DOI: 10.34133/bmef.0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/30/2023] [Indexed: 10/19/2023] Open
Abstract
In the era of personalized oncology, there have been accelerated efforts to develop clinically relevant platforms to test drug sensitivities of individual cancers. An ideal assay will serve as a diagnostic companion to inform the oncologist of the various treatments that are sensitive and insensitive, thus improving outcome while minimizing unnecessary toxicities and costs. To date, no such platform exists for clinical use, but promising approaches are on the horizon that take advantage of improved techniques in creating human cancer models that encompass the entire tumor microenvironment, alongside technologies for assessing and analyzing tumor response. This review summarizes a number of current strategies that make use of intact human cancer tissues as organotypic cultures in drug sensitivity testing.
Collapse
Affiliation(s)
- Yu Ling Huang
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Heidi Kenerson
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Xiuyun Jiang
- Department of Surgery, University of Washington, Seattle, WA, USA
| | | | - Qiang Tian
- National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leroy Hood
- Institute for Systems Biology, Phenome Health Institute, Seattle, WA, USA
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, WA, USA
| |
Collapse
|
13
|
Ghorbaninejad M, Asadzadeh-Aghdaei H, Baharvand H, Meyfour A. Intestinal organoids: A versatile platform for modeling gastrointestinal diseases and monitoring epigenetic alterations. Life Sci 2023; 319:121506. [PMID: 36858311 DOI: 10.1016/j.lfs.2023.121506] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/03/2023]
Abstract
Considering the significant limitations of conventional 2D cell cultures and tissue in vitro models, creating intestinal organoids has burgeoned as an ideal option to recapitulate the heterogeneity of the native intestinal epithelium. Intestinal organoids can be developed from either tissue-resident adult stem cells (ADSs) or pluripotent stem cells (PSCs) in both forms induced PSCs and embryonic stem cells. Here, we review current advances in the development of intestinal organoids that have led to a better recapitulation of the complexity, physiology, morphology, function, and microenvironment of the intestine. We discuss current applications of intestinal organoids with an emphasis on disease modeling. In particular, we point out recent studies on SARS-CoV-2 infection in human intestinal organoids. We also discuss the less explored application of intestinal organoids in epigenetics by highlighting the role of epigenetic modifications in intestinal development, homeostasis, and diseases, and subsequently the power of organoids in mirroring the regulatory role of epigenetic mechanisms in these conditions and introducing novel predictive/diagnostic biomarkers. Finally, we propose 3D organoid models to evaluate the effects of novel epigenetic drugs (epi-drugs) on the treatment of GI diseases where epigenetic mechanisms play a key role in disease development and progression, particularly in colorectal cancer treatment and epigenetically acquired drug resistance.
Collapse
Affiliation(s)
- Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh-Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Baharvand
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
14
|
Mohandas S, Gayatri V, Kumaran K, Gopinath V, Paulmurugan R, Ramkumar KM. New Frontiers in Three-Dimensional Culture Platforms to Improve Diabetes Research. Pharmaceutics 2023; 15:pharmaceutics15030725. [PMID: 36986591 PMCID: PMC10056755 DOI: 10.3390/pharmaceutics15030725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Diabetes mellitus is associated with defects in islet β-cell functioning and consequent hyperglycemia resulting in multi-organ damage. Physiologically relevant models that mimic human diabetic progression are urgently needed to identify new drug targets. Three-dimensional (3D) cell-culture systems are gaining a considerable interest in diabetic disease modelling and are being utilized as platforms for diabetic drug discovery and pancreatic tissue engineering. Three-dimensional models offer a marked advantage in obtaining physiologically relevant information and improve drug selectivity over conventional 2D (two-dimensional) cultures and rodent models. Indeed, recent evidence persuasively supports the adoption of appropriate 3D cell technology in β-cell cultivation. This review article provides a considerably updated view of the benefits of employing 3D models in the experimental workflow compared to conventional animal and 2D models. We compile the latest innovations in this field and discuss the various strategies used to generate 3D culture models in diabetic research. We also critically review the advantages and the limitations of each 3D technology, with particular attention to the maintenance of β-cell morphology, functionality, and intercellular crosstalk. Furthermore, we emphasize the scope of improvement needed in the 3D culture systems employed in diabetes research and the promises they hold as excellent research platforms in managing diabetes.
Collapse
Affiliation(s)
- Sundhar Mohandas
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vijaya Gayatri
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Kriya Kumaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vipin Gopinath
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Molecular Oncology Division, Malabar Cancer Centre, Moozhikkara P.O, Thalassery 670103, Kerala, India
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| |
Collapse
|
15
|
Cao UMN, Zhang Y, Chen J, Sayson D, Pillai S, Tran SD. Microfluidic Organ-on-A-chip: A Guide to Biomaterial Choice and Fabrication. Int J Mol Sci 2023; 24:3232. [PMID: 36834645 PMCID: PMC9966054 DOI: 10.3390/ijms24043232] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Organ-on-A-chip (OoAC) devices are miniaturized, functional, in vitro constructs that aim to recapitulate the in vivo physiology of an organ using different cell types and extracellular matrix, while maintaining the chemical and mechanical properties of the surrounding microenvironments. From an end-point perspective, the success of a microfluidic OoAC relies mainly on the type of biomaterial and the fabrication strategy employed. Certain biomaterials, such as PDMS (polydimethylsiloxane), are preferred over others due to their ease of fabrication and proven success in modelling complex organ systems. However, the inherent nature of human microtissues to respond differently to surrounding stimulations has led to the combination of biomaterials ranging from simple PDMS chips to 3D-printed polymers coated with natural and synthetic materials, including hydrogels. In addition, recent advances in 3D printing and bioprinting techniques have led to the powerful combination of utilizing these materials to develop microfluidic OoAC devices. In this narrative review, we evaluate the different materials used to fabricate microfluidic OoAC devices while outlining their pros and cons in different organ systems. A note on combining the advances made in additive manufacturing (AM) techniques for the microfabrication of these complex systems is also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
16
|
Dashtaki ME, Ghasemi S. CRISPR/Cas9-based Gene Therapies for Fighting Drug Resistance Mediated by Cancer Stem Cells. Curr Gene Ther 2023; 23:41-50. [PMID: 36056851 DOI: 10.2174/1566523222666220831161225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/11/2022] [Accepted: 06/11/2022] [Indexed: 02/08/2023]
Abstract
Cancer stem cells (CSCs) are cancer-initiating cells found in most tumors and hematological cancers. CSCs are involved in cells progression, recurrence of tumors, and drug resistance. Current therapies have been focused on treating the mass of tumor cells and cannot eradicate the CSCs. CSCs drug-specific targeting is considered as an approach to precisely target these cells. Clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) gene-editing systems are making progress and showing promise in the cancer research field. One of the attractive applications of CRISPR/Cas9 as one approach of gene therapy is targeting the critical genes involved in drug resistance and maintenance of CSCs. The synergistic effects of gene editing as a novel gene therapy approach and traditional therapeutic methods, including chemotherapy, can resolve drug resistance challenges and regression of the cancers. This review article considers different aspects of CRISPR/Cas9 ability in the study and targeting of CSCs with the intention to investigate their application in drug resistance.
Collapse
Affiliation(s)
- Masoumeh Eliyasi Dashtaki
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
17
|
Yun C, Kim SH, Jung YS. Current Research Trends in the Application of In Vitro Three-Dimensional Models of Liver Cells. Pharmaceutics 2022; 15:pharmaceutics15010054. [PMID: 36678683 PMCID: PMC9866911 DOI: 10.3390/pharmaceutics15010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The liver produces and stores various nutrients that are necessary for the body and serves as a chemical plant, metabolizing carbohydrates, fats, hormones, vitamins, and minerals. It is also a vital organ for detoxifying drugs and exogenous harmful substances. Culturing liver cells in vitro under three-dimensional (3D) conditions is considered a primary mechanism for liver tissue engineering. The 3D cell culture system is designed to allow cells to interact in an artificially created environment and has the advantage of mimicking the physiological characteristics of cells in vivo. This system facilitates contact between the cells and the extracellular matrix. Several technically different approaches have been proposed, including bioreactors, chips, and plate-based systems in fluid or static media composed of chemically diverse materials. Compared to conventional two-dimensional monolayer culture in vitro models, the ability to predict the function of the tissues, including the drug metabolism and chemical toxicity, has been enhanced by developing three-dimensional liver culture models. This review discussed the methodology of 3D cell cultures and summarized the advantages of an in vitro liver platform using 3D culture technology.
Collapse
|
18
|
Lim S, Kim SW, Kim IK, Song BW, Lee S. Organ-on-a-chip: Its use in cardiovascular research. Clin Hemorheol Microcirc 2022; 83:315-339. [DOI: 10.3233/ch-221428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Organ-on-a-chip (OOAC) has attracted great attention during the last decade as a revolutionary alternative to conventional animal models. This cutting-edge technology has also brought constructive changes to the field of cardiovascular research. The cardiovascular system, especially the heart as a well-protected vital organ, is virtually impossible to replicate in vitro with conventional approaches. This made scientists assume that they needed to use animal models for cardiovascular research. However, the frequent failure of animal models to correctly reflect the native cardiovascular system necessitated a search for alternative platforms for preclinical studies. Hence, as a promising alternative to conventional animal models, OOAC technology is being actively developed and tested in a wide range of biomedical fields, including cardiovascular research. Therefore, in this review, the current literature on the use of OOACs for cardiovascular research is presented with a focus on the basis for using OOACs, and what has been specifically achieved by using OOACs is also discussed. By providing an overview of the current status of OOACs in cardiovascular research and its future perspectives, we hope that this review can help to develop better and optimized research strategies for cardiovascular diseases (CVDs) as well as identify novel applications of OOACs in the near future.
Collapse
Affiliation(s)
- Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| |
Collapse
|
19
|
Szostak B, Gorący A, Pala B, Rosik J, Ustianowski Ł, Pawlik A. Latest models for the discovery and development of rheumatoid arthritis drugs. Expert Opin Drug Discov 2022; 17:1261-1278. [PMID: 36184990 DOI: 10.1080/17460441.2022.2131765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic autoimmune disease that reduces the quality of life. The current speed of development of therapeutic agents against RA is not satisfactory. Models on which initial experiments are conducted do not fully reflect human pathogenesis. Overcoming this oversimplification might be a crucial step to accelerate studies on RA treatment. AREAS COVERED The current approaches to produce novel models or to improve currently available models for the development of RA drugs have been discussed. Advantages and drawbacks of two- and three-dimensional cell cultures and animal models have been described based on recently published results of the studies. Moreover, approaches such as tissue engineering or organ-on-a-chip have been reviewed. EXPERT OPINION The cell cultures and animal models used to date appear to be of limited value due to the complexity of the processes involved in RA. Current models in RA research should take into account the heterogeneity of patients in terms of disease subtypes, course, and activity. Several advanced models and tools using human cells and tissues have been developed, including three-dimensional tissues, liquid bioreactors, and more complex joint-on-a-chip devices. This may increase knowledge of the molecular mechanisms leading to disease development, to help identify new biomarkers for early detection, and to develop preventive strategies and more effective treatments.
Collapse
Affiliation(s)
- Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Anna Gorący
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Bartłomiej Pala
- Department of Neurosurgery, Pomeranian Medical University Hospital No. 1, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland.,Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Łukasz Ustianowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
20
|
Franca CM, Balbinot GDS, Cunha D, Saboia VDPA, Ferracane J, Bertassoni LE. In-vitro models of biocompatibility testing for restorative dental materials: From 2D cultures to organs on-a-chip. Acta Biomater 2022; 150:58-66. [PMID: 35933103 PMCID: PMC9814917 DOI: 10.1016/j.actbio.2022.07.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/13/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023]
Abstract
Dental caries is a biofilm-mediated, diet-modulated, multifactorial and dynamic disease that affects more than 90% of adults in Western countries. The current treatment for decayed tissue is based on using materials to replace the lost enamel or dentin. More than 500 million dental restorations are placed annually worldwide, and materials used for these purposes either directly or indirectly interact with dentin and pulp tissues. The development and understanding of the effects of restorative dental materials are based on different in-vitro and in-vivo tests, which have been evolving with time. In this review, we first discuss the characteristics of the tooth and the dentin-pulp interface that are unique for materials testing. Subsequently, we discuss frequently used in-vitro tests to evaluate the biocompatibility of dental materials commonly used for restorative procedures. Finally, we present our perspective on the future directions for biological research on dental materials using tissue engineering and organs on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Dental caries is still the most prevalent infectious disease globally, requiring more than 500 million restorations to be placed every year. Regrettably, the failure rates of such restorations are still high. Those rates are partially based on the fact that current platforms to test dental materials are somewhat inaccurate in reproducing critical components of the complex oral microenvironment. Thus, there is a collective effort to develop new materials while evolving the platforms to test them. In this context, the present review critically discusses in-vitro models used to evaluate the biocompatibility of restorative dental materials and brings a perspective on future directions for tissue-engineered and organs-on-a-chip platforms for testing new dental materials.
Collapse
Affiliation(s)
- Cristiane Miranda Franca
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Gabriela de Souza Balbinot
- Dental Materials Laboratory, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Diana Cunha
- Post-Graduation Program in Dentistry, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Jack Ferracane
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Luiz E Bertassoni
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States; Center for Regenerative Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, United States; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States; Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, United States.
| |
Collapse
|
21
|
Nahak BK, Mishra A, Preetam S, Tiwari A. Advances in Organ-on-a-Chip Materials and Devices. ACS APPLIED BIO MATERIALS 2022; 5:3576-3607. [PMID: 35839513 DOI: 10.1021/acsabm.2c00041] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The organ-on-a-chip (OoC) paves a way for biomedical applications ranging from preclinical to clinical translational precision. The current trends in the in vitro modeling is to reduce the complexity of human organ anatomy to the fundamental cellular microanatomy as an alternative of recreating the entire cell milieu that allows systematic analysis of medicinal absorption of compounds, metabolism, and mechanistic investigation. The OoC devices accurately represent human physiology in vitro; however, it is vital to choose the correct chip materials. The potential chip materials include inorganic, elastomeric, thermoplastic, natural, and hybrid materials. Despite the fact that polydimethylsiloxane is the most commonly utilized polymer for OoC and microphysiological systems, substitute materials have been continuously developed for its advanced applications. The evaluation of human physiological status can help to demonstrate using noninvasive OoC materials in real-time procedures. Therefore, this Review examines the materials used for fabricating OoC devices, the application-oriented pros and cons, possessions for device fabrication and biocompatibility, as well as their potential for downstream biochemical surface alteration and commercialization. The convergence of emerging approaches, such as advanced materials, artificial intelligence, machine learning, three-dimensional (3D) bioprinting, and genomics, have the potential to perform OoC technology at next generation. Thus, OoC technologies provide easy and precise methodologies in cost-effective clinical monitoring and treatment using standardized protocols, at even personalized levels. Because of the inherent utilization of the integrated materials, employing the OoC with biomedical approaches will be a promising methodology in the healthcare industry.
Collapse
Affiliation(s)
- Bishal Kumar Nahak
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Anshuman Mishra
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| |
Collapse
|
22
|
Eastman AJ, Noble KN, Pensabene V, Aronoff DM. Leveraging bioengineering to assess cellular functions and communication within human fetal membranes. J Matern Fetal Neonatal Med 2022; 35:2795-2807. [PMID: 32787482 PMCID: PMC7878582 DOI: 10.1080/14767058.2020.1802716] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The fetal membranes enclose the growing fetus and amniotic fluid. Preterm prelabor rupture of fetal membranes is a leading cause of preterm birth. Fetal membranes are composed of many different cell types, both structural and immune. These cells must coordinate functions for tensile strength and membrane integrity to contain the growing fetus and amniotic fluid. They must also balance immune responses to pathogens with maintaining maternal-fetal tolerance. Perturbation of this equilibrium can lead to preterm premature rupture of membranes without labor. In this review, we describe the formation of the fetal membranes to orient the reader, discuss some of the common forms of communication between the cell types of the fetal membranes, and delve into the methods used to tease apart this paracrine signaling within the membranes, including emerging technologies such as organ-on-chip models of membrane immunobiology.
Collapse
Affiliation(s)
- Alison J. Eastman
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kristen N. Noble
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, TN 37202 USA
| | - Virginia Pensabene
- School of Electronic and Electrical Engineering, University of Leeds, Leeds, UK,School of Medicine, Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - David M. Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Corresponding author: David M. Aronoff, MD, 1161 21st Ave South, A-2200 MCN, Nashville, TN 37232-2582, (615) 322-8972 (tel),
| |
Collapse
|
23
|
Varzideh F, Mone P, Santulli G. Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:168. [PMID: 35447728 PMCID: PMC9028595 DOI: 10.3390/bioengineering9040168] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be used to generate various cell types in the human body. Hence, hiPSC-derived cardiomyocytes (hiPSC-CMs) represent a significant cell source for disease modeling, drug testing, and regenerative medicine. The immaturity of hiPSC-CMs in two-dimensional (2D) culture limit their applications. Cardiac tissue engineering provides a new promise for both basic and clinical research. Advanced bioengineered cardiac in vitro models can create contractile structures that serve as exquisite in vitro heart microtissues for drug testing and disease modeling, thereby promoting the identification of better treatments for cardiovascular disorders. In this review, we will introduce recent advances of bioengineering technologies to produce in vitro cardiac tissues derived from hiPSCs.
Collapse
Affiliation(s)
- Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
24
|
Vivas A, van den Berg A, Passier R, Odijk M, van der Meer AD. Fluidic circuit board with modular sensor and valves enables stand-alone, tubeless microfluidic flow control in organs-on-chips. LAB ON A CHIP 2022; 22:1231-1243. [PMID: 35178541 PMCID: PMC8922413 DOI: 10.1039/d1lc00999k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/09/2022] [Indexed: 05/13/2023]
Abstract
Organs-on-chips are a unique class of microfluidic in vitro cell culture models, in which the in vivo tissue microenvironment is mimicked. Unfortunately, their widespread use is hampered by their operation complexity and incompatibility with end-user research settings. To address these issues, many commercial and non-commercial platforms have been developed for semi-automated culture of organs-on-chips. However, these organ-on-chip culture platforms each represent a closed ecosystem, with very little opportunity to interchange and integrate components from different platforms or to develop new ones. The translational organ-on-chip platform (TOP) is a multi-institutional effort to develop an open platform for automated organ-on-chip culture and integration of components from various developers. Central to TOP is the fluidic circuit board (FCB), a microfluidic plate with the form factor of a typical well plate. The FCB enables microfluidic control of multiple components like sensors or organ-on-chip devices through an interface based on openly available standards. Here, we report an FCB to integrate commercial and in-house developed components forming a stand-alone flow control system for organs-on-chips. The control system is able to achieve constant and pulsatile flow recirculation through a connected organ-on-chip device. We demonstrate that this system is able to automatically perfuse a heart-on-chip device containing co-cultures of cardiac tissues derived from human pluripotent stem cell-derived cardiomyocytes and monolayers of endothelial cells for five days. Altogether, we conclude that open technology platforms allow the integration of components from different sources to form functional and fit-for-purpose organ-on-chip systems. We anticipate that open platforms will play a central role in catalyzing and maturing further technological development of organ-on-chip culture systems.
Collapse
Affiliation(s)
- Aisen Vivas
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, Enschede 7500 AE, The Netherlands.
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Albert van den Berg
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Robert Passier
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, Enschede 7500 AE, The Netherlands.
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Mathieu Odijk
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7500 AE, The Netherlands
| | - Andries D van der Meer
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, Enschede 7500 AE, The Netherlands.
| |
Collapse
|
25
|
Antisense RNA Therapeutics: A Brief Overview. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2434:33-49. [PMID: 35213008 DOI: 10.1007/978-1-0716-2010-6_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nucleic acid therapeutics is a growing field aiming to treat human conditions that has gained special attention due to the successful development of mRNA vaccines against SARS-CoV-2. Another type of nucleic acid therapeutics is antisense oligonucleotides, versatile tools that can be used in multiple ways to target pre-mRNA and mRNA. While some years ago these molecules were just considered a useful research tool and a curiosity in the clinical market, this has rapidly changed. These molecules are promising strategies for personalized treatments for rare genetic diseases and they are in development for very common disorders too. In this chapter, we provide a brief description of the different mechanisms of action of these RNA therapeutic molecules, with clear examples at preclinical and clinical stages.
Collapse
|
26
|
Barker S, Dagys L, Hale W, Ripka B, Eills J, Sharma M, Levitt MH, Utz M. Direct Production of a Hyperpolarized Metabolite on a Microfluidic Chip. Anal Chem 2022; 94:3260-3267. [PMID: 35147413 PMCID: PMC9096798 DOI: 10.1021/acs.analchem.1c05030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/24/2022] [Indexed: 12/28/2022]
Abstract
Microfluidic systems hold great potential for the study of live microscopic cultures of cells, tissue samples, and small organisms. Integration of hyperpolarization would enable quantitative studies of metabolism in such volume limited systems by high-resolution NMR spectroscopy. We demonstrate, for the first time, the integrated generation and detection of a hyperpolarized metabolite on a microfluidic chip. The metabolite [1-13C]fumarate is produced in a nuclear hyperpolarized form by (i) introducing para-enriched hydrogen into the solution by diffusion through a polymer membrane, (ii) reaction with a substrate in the presence of a ruthenium-based catalyst, and (iii) conversion of the singlet-polarized reaction product into a magnetized form by the application of a radiofrequency pulse sequence, all on the same microfluidic chip. The microfluidic device delivers a continuous flow of hyperpolarized material at the 2.5 μL/min scale, with a polarization level of 4%. We demonstrate two methods for mitigating singlet-triplet mixing effects which otherwise reduce the achieved polarization level.
Collapse
Affiliation(s)
- Sylwia
J. Barker
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Laurynas Dagys
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - William Hale
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Department
of Chemistry, University of Florida, Gainesville 32611, United States
| | - Barbara Ripka
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - James Eills
- Institute
for Physics, Johannes Gutenberg University, D-55090 Mainz, Germany
- GSI
Helmholtzzentrum für Schwerionenforschung GmbH, Helmholtz-Institut Mainz, 55128 Mainz, Germany
| | - Manvendra Sharma
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Malcolm H. Levitt
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Marcel Utz
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
27
|
Kabay G, Manz A, Dincer C. Microfluidic Roadmap for Translational Nanotheranostics. SMALL METHODS 2022; 6:e2101217. [PMID: 34957704 DOI: 10.1002/smtd.202101217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/22/2021] [Indexed: 06/14/2023]
Abstract
Nanotheranostic materials (NTMs) shed light on the mechanisms responsible for complex diseases such as cancer because they enable making a diagnosis, monitoring the disease progression, and applying a targeted therapy simultaneously. However, several issues such as the reproducibility and mass production of NTMs hamper their application for clinical practice. To address these issues and facilitate the clinical application of NTMs, microfluidic systems have been increasingly used. This perspective provides a glimpse into the current state-of-art of NTM research, emphasizing the methods currently employed at each development stage of NTMs and the related open problems. This work reviews microfluidic technologies used to develop NTMs, ranging from the fabrication and testing of a single NTM up to their manufacturing on a large scale. Ultimately, a step-by-step vision on the future development of NTMs for clinical practice enabled by microfluidics techniques is provided.
Collapse
Affiliation(s)
- Gozde Kabay
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110, Freiburg, Germany
- University of Freiburg, FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, 79110, Freiburg, Germany
| | - Andreas Manz
- Korea Institute of Science and Technology (KIST) in Europe, 66123, Saarbrücken, Germany
| | - Can Dincer
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110, Freiburg, Germany
- University of Freiburg, FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, 79110, Freiburg, Germany
| |
Collapse
|
28
|
Fanizza F, Campanile M, Forloni G, Giordano C, Albani D. Induced pluripotent stem cell-based organ-on-a-chip as personalized drug screening tools: A focus on neurodegenerative disorders. J Tissue Eng 2022; 13:20417314221095339. [PMID: 35570845 PMCID: PMC9092580 DOI: 10.1177/20417314221095339] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/04/2022] [Indexed: 01/15/2023] Open
Abstract
The Organ-on-a-Chip (OoC) technology shows great potential to revolutionize the drugs development pipeline by mimicking the physiological environment and functions of human organs. The translational value of OoC is further enhanced when combined with patient-specific induced pluripotent stem cells (iPSCs) to develop more realistic disease models, paving the way for the development of a new generation of patient-on-a-chip devices. iPSCs differentiation capacity leads to invaluable improvements in personalized medicine. Moreover, the connection of single-OoC into multi-OoC or body-on-a-chip allows to investigate drug pharmacodynamic and pharmacokinetics through the study of multi-organs cross-talks. The need of a breakthrough thanks to this technology is particularly relevant within the field of neurodegenerative diseases, where the number of patients is increasing and the successful rate in drug discovery is worryingly low. In this review we discuss current iPSC-based OoC as drug screening models and their implication in development of new therapies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fanizza
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Marzia Campanile
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
29
|
Moreira A, Müller M, Costa PF, Kohl Y. Advanced In Vitro Lung Models for Drug and Toxicity Screening: The Promising Role of Induced Pluripotent Stem Cells. Adv Biol (Weinh) 2021; 6:e2101139. [PMID: 34962104 DOI: 10.1002/adbi.202101139] [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: 08/23/2021] [Revised: 11/25/2021] [Indexed: 12/24/2022]
Abstract
The substantial socioeconomic burden of lung diseases, recently highlighted by the disastrous impact of the coronavirus disease 2019 (COVID-19) pandemic, accentuates the need for interventive treatments capable of decelerating disease progression, limiting organ damage, and contributing to a functional tissue recovery. However, this is hampered by the lack of accurate human lung research models, which currently fail to reproduce the human pulmonary architecture and biochemical environment. Induced pluripotent stem cells (iPSCs) and organ-on-chip (OOC) technologies possess suitable characteristics for the generation of physiologically relevant in vitro lung models, allowing for developmental studies, disease modeling, and toxicological screening. Importantly, these platforms represent potential alternatives for animal testing, according to the 3Rs (replace, reduce, refine) principle, and hold promise for the identification and approval of new chemicals under the European REACH (registration, evaluation, authorization and restriction of chemicals) framework. As such, this review aims to summarize recent progress made in human iPSC- and OOC-based in vitro lung models. A general overview of the present applications of in vitro lung models is presented, followed by a summary of currently used protocols to generate different lung cell types from iPSCs. Lastly, recently developed iPSC-based lung models are discussed.
Collapse
Affiliation(s)
| | - Michelle Müller
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Pedro F Costa
- BIOFABICS, Rua Alfredo Allen 455, Porto, 4200-135, Portugal
| | - Yvonne Kohl
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany.,Postgraduate Course for Toxicology and Environmental Toxicology, Medical Faculty, University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| |
Collapse
|
30
|
Lee SY, Kim D, Lee SH, Sung JH. Microtechnology-based in vitro models: Mimicking liver function and pathophysiology. APL Bioeng 2021; 5:041505. [PMID: 34703969 PMCID: PMC8520487 DOI: 10.1063/5.0061896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
The liver plays important roles in drug metabolism and homeostasis. The metabolism and biotransformation can not only affect the efficacy of drugs but also result in hepatotoxicity and drug-induced liver injury. Understanding the complex physiology of the liver and the pathogenetic mechanisms of liver diseases is essential for drug development. Conventional in vitro models have limitations in the ability to predict drug effects, due to the lack of physiological relevance. Recently, the liver-on-a-chip platform has been developed to reproduce the microarchitecture and in vivo environment of the liver. These efforts have improved the physiological relevance of the liver tissue used in the platform and have demonstrated its applicability to drug screening and disease models. In this review, we summarize the recent development of liver-on-a-chip models that closely mimic the in vivo liver environments and liver diseases.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| | - Donghyun Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, South Korea
| | - Seung Hwan Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, South Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| |
Collapse
|
31
|
Recent Advances in Chronotherapy Targeting Respiratory Diseases. Pharmaceutics 2021; 13:pharmaceutics13122008. [PMID: 34959290 PMCID: PMC8704788 DOI: 10.3390/pharmaceutics13122008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 11/17/2022] Open
Abstract
Respiratory diseases contribute to a significant percentage of mortality and morbidity worldwide. The circadian rhythm is a natural biological process where our bodily functions align with the 24 h oscillation (sleep-wake cycle) process and are controlled by the circadian clock protein/gene. Disruption of the circadian rhythm could alter normal lung function. Chronotherapy is a type of therapy provided at specific time intervals based on an individual's circadian rhythm. This would allow the drug to show optimum action, and thereby modulate its pharmacokinetics to lessen unwanted or unintended effects. In this review, we deliberated on the recent advances employed in chrono-targeted therapeutics for chronic respiratory diseases.
Collapse
|
32
|
Bakhchova L, Jantaree P, Gupta A, Isermann B, Steinmann U, Naumann M. On-a-Chip-Based Sensitive Detection of Drug-Induced Apoptosis in Polarized Gastric Epithelial Cells. ACS Biomater Sci Eng 2021; 7:5474-5483. [PMID: 34704732 DOI: 10.1021/acsbiomaterials.1c01094] [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] [Indexed: 01/22/2023]
Abstract
Microfluidic devices for culturing cells have been successfully utilized for biomedical applications, including drug screening. Several cell lines could be cultivated in microengineered environments with promising results, but gastric cell lines have not yet been widely used or studied. Therefore, this study focuses on establishing a polarized gastric epithelial monolayer on-a-chip and describes a general-purpose methodology applicable for bonding any porous material to PDMS through an adhesive sublayer. The fully transparent microfluidic chip consists of two microfluidic channels separated by a collagen-coated porous membrane and lined by human polarized gastric epithelial (NCI-N87) cells. We present considerations on how to ensure continuous and stable flow through the channels. The continuous flow rate was achieved using a pressure-driven pump. Media flow at a constant rate (0.5 μL/min) rapidly led the gastric epithelial cells to develop into a polarized monolayer. The barrier integrity was assessed by the FITC-dextran test. The generation of a monolayer was faster than in the static Boyden chamber. Moreover, fluorescence microscopy was used to monitor the apoptotic cell death of gastric epithelial monolayers on-a-chip in response to camptothecin, a therapeutic gastric cancer drug.
Collapse
Affiliation(s)
- Liubov Bakhchova
- Institute for Automation Technology, Otto von Guericke University, Universitätsplatz 2, Magdeburg 39106, Germany.,Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Leipziger Straße 44, Magdeburg 39120, Germany
| | - Phatcharida Jantaree
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Leipziger Straße 44, Magdeburg 39120, Germany
| | - Anubhuti Gupta
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Leipziger Straße 44, Magdeburg 39120, Germany.,Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, Universitätsklinikum Leipzig, Paul-List-Straße 13-15, Leipzig 04103, Germany
| | - Berend Isermann
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, Universitätsklinikum Leipzig, Paul-List-Straße 13-15, Leipzig 04103, Germany
| | - Ulrike Steinmann
- Institute for Automation Technology, Otto von Guericke University, Universitätsplatz 2, Magdeburg 39106, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Leipziger Straße 44, Magdeburg 39120, Germany
| |
Collapse
|
33
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
34
|
Oliveira IM, Carvalho MR, Fernandes DC, Abreu CM, Maia FR, Pereira H, Caballero D, Kundu SC, Reis RL, Oliveira JM. Modulation of inflammation by anti-TNF α mAb-dendrimer nanoparticles loaded in tyramine-modified gellan gum hydrogels in a cartilage-on-a-chip model. J Mater Chem B 2021; 9:4211-4218. [PMID: 33998627 DOI: 10.1039/d1tb00802a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune and chronic inflammatory disease characterized by joint inflammation. Since the inflammatory condition plays an important role in the disease process, it is important to develop and test new therapeutic approaches that specifically target and treat joint inflammation. In this study, a human 3D inflammatory cartilage-on-a-chip model was established to test the therapeutic efficacy of anti-TNFα mAb-CS/PAMAM dendrimer NPs loaded-Tyramine-Gellan Gum in the treatment of inflammation. The results showed that the proposed therapeutic approach applied to the human monocyte cell line (THP-1) and human chondrogenic primary cells (hCH) cell-based inflammation system revealed an anti-inflammatory capacity that increased over 14 days. It was also possible to observe that Coll type II was highly expressed by inflamed hCH upon the culture with anti-TNF α mAb-CS/PAMAM dendrimer NPs, indicating that the hCH cells were able maintain their biological function. The developed preclinical model allowed us to provide more robust data on the potential therapeutic effect of anti-TNF α mAb-CS/PAMAM dendrimer NPs loaded-Ty-GG hydrogel in a physiologically relevant model.
Collapse
Affiliation(s)
- I M Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - M R Carvalho
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - D C Fernandes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - C M Abreu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - F R Maia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - H Pereira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal and Orthopedic Department, Povoa de Varzim - Vila do Conde Hospital Centre, Portugal and Ripoll & De Prado Sport Clinic, Spain
| | - D Caballero
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - S C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - R L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J M Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
35
|
Costa J, Mackay R, de Aguiar Greca SC, Corti A, Silva E, Karteris E, Ahluwalia A. The Role of the 3Rs for Understanding and Modeling the Human Placenta. J Clin Med 2021; 10:jcm10153444. [PMID: 34362227 PMCID: PMC8347836 DOI: 10.3390/jcm10153444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Modeling the physiology of the human placenta is still a challenge, despite the great number of scientific advancements made in the field. Animal models cannot fully replicate the structure and function of the human placenta and pose ethical and financial hurdles. In addition, increasingly stricter animal welfare legislation worldwide is incentivizing the use of 3R (reduction, refinement, replacement) practices. What efforts have been made to develop alternative models for the placenta so far? How effective are they? How can we improve them to make them more predictive of human pathophysiology? To address these questions, this review aims at presenting and discussing the current models used to study phenomena at the placenta level: in vivo, ex vivo, in vitro and in silico. We describe the main achievements and opportunities for improvement of each type of model and critically assess their individual and collective impact on the pursuit of predictive studies of the placenta in line with the 3Rs and European legislation.
Collapse
Affiliation(s)
- Joana Costa
- Centro di Ricerca E.Piaggio, University of Pisa, 56126 Pisa, Italy; (J.C.); (A.C.)
| | - Ruth Mackay
- Centre for Genome Engineering and Maintenance, Department of Mechanical and Aerospace Engineering, Brunel University London, Uxbridge UB8 3PH, UK;
| | | | - Alessandro Corti
- Centro di Ricerca E.Piaggio, University of Pisa, 56126 Pisa, Italy; (J.C.); (A.C.)
- Department of Translational Medicine, University of Pisa, 56126 Pisa, Italy
| | - Elisabete Silva
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (S.-C.d.A.G.); (E.S.); (E.K.)
| | - Emmanouil Karteris
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (S.-C.d.A.G.); (E.S.); (E.K.)
| | - Arti Ahluwalia
- Centro di Ricerca E.Piaggio, University of Pisa, 56126 Pisa, Italy; (J.C.); (A.C.)
- Department of Information Engineering, University of Pisa, 56122 Pisa, Italy
- Interuniversity Centro for the Promotion of 3Rs Principles in Teaching and Research (Centro3R), Italy
- Correspondence:
| |
Collapse
|
36
|
Kulsharova G, Kurmangaliyeva A. Liver microphysiological platforms for drug metabolism applications. Cell Prolif 2021; 54:e13099. [PMID: 34291515 PMCID: PMC8450120 DOI: 10.1111/cpr.13099] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/21/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Drug development is a costly and lengthy process with low success rates. To improve the efficiency of drug development, there has been an increasing need in developing alternative methods able to eliminate toxic compounds early in the drug development pipeline. Drug metabolism plays a key role in determining the efficacy of a drug and its potential side effects. Since drug metabolism occurs mainly in the liver, liver cell‐based alternative engineering platforms have been growing in the last decade. Microphysiological liver cell‐based systems called liver‐on‐a‐chip platforms can better recapitulate the environment for human liver cells in laboratory settings and have the potential to reduce the number of animal models used in drug development by predicting the response of the liver to a drug in vitro. In this review, we discuss the liver microphysiological platforms from the perspective of drug metabolism studies. We highlight the stand‐alone liver‐on‐a‐chip platforms and multi‐organ systems integrating liver‐on‐a‐chip devices used for drug metabolism mimicry in vitro and review the state‐of‐the‐art platforms reported in the last few years. With the development of more robust and reproducible liver cell‐based microphysiological platforms, the drug development field has the potential of reducing the costs and lengths associated with currently existing drug testing methods.
Collapse
Affiliation(s)
- Gulsim Kulsharova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | | |
Collapse
|
37
|
Wang L, Zhou MB, Zhang H. The Emerging Role of Topical Ocular Drugs to Target the Posterior Eye. Ophthalmol Ther 2021; 10:465-494. [PMID: 34218424 PMCID: PMC8319259 DOI: 10.1007/s40123-021-00365-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of chronic fundus diseases is increasing with the aging of the general population. The treatment of these intraocular diseases relies on invasive drug delivery because of the globular structure and multiple barriers of the eye. Frequent intraocular injections bring heavy burdens to the medical care system and patients. The use of topical drugs to treat retinal diseases has always been an attractive solution. The fast development of new materials and technologies brings the possibility to develop innovative topical formulations. This article reviews anatomical and physiological barriers of the eye which affect the bioavailability of topical drugs. In addition, we summarize innovative topical formulations which enhance the permeability of drugs through the ocular surface and/or extend the drug retention time in the eye. This article also reviews the differences of eyes between different laboratory animals to address the translational challenges of preclinical models. The fast development of in vitro eye models may provide more tools to increase the clinical translationality of topical formulations for intraocular diseases. Clinical successes of topical formulations rely on continuous and collaborative efforts between different disciplines.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hui Zhang
- Yuanpu Eye Biopharmaceutical Co. Ltd., Chengdu, China.
- , No. 14 Jiuxing Avenue, Gaoxin District, Chengdu, China.
| |
Collapse
|
38
|
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: 33] [Impact Index Per Article: 11.0] [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
|
39
|
Zheng F, Xiao Y, Liu H, Fan Y, Dao M. Patient-Specific Organoid and Organ-on-a-Chip: 3D Cell-Culture Meets 3D Printing and Numerical Simulation. Adv Biol (Weinh) 2021; 5:e2000024. [PMID: 33856745 PMCID: PMC8243895 DOI: 10.1002/adbi.202000024] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/13/2021] [Indexed: 12/11/2022]
Abstract
The last few decades have witnessed diversified in vitro models to recapitulate the architecture and function of living organs or tissues and contribute immensely to advances in life science. Two novel 3D cell culture models: 1) Organoid, promoted mainly by the developments of stem cell biology and 2) Organ-on-a-chip, enhanced primarily due to microfluidic technology, have emerged as two promising approaches to advance the understanding of basic biological principles and clinical treatments. This review describes the comparable distinct differences between these two models and provides more insights into their complementarity and integration to recognize their merits and limitations for applicable fields. The convergence of the two approaches to produce multi-organoid-on-a-chip or human organoid-on-a-chip is emerging as a new approach for building 3D models with higher physiological relevance. Furthermore, rapid advancements in 3D printing and numerical simulations, which facilitate the design, manufacture, and results-translation of 3D cell culture models, can also serve as novel tools to promote the development and propagation of organoid and organ-on-a-chip systems. Current technological challenges and limitations, as well as expert recommendations and future solutions to address the promising combinations by incorporating organoids, organ-on-a-chip, 3D printing, and numerical simulation, are also summarized.
Collapse
Affiliation(s)
- Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Yuminghao Xiao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
40
|
Carvalho V, Gonçalves I, Lage T, Rodrigues RO, Minas G, Teixeira SFCF, Moita AS, Hori T, Kaji H, Lima RA. 3D Printing Techniques and Their Applications to Organ-on-a-Chip Platforms: A Systematic Review. SENSORS (BASEL, SWITZERLAND) 2021; 21:3304. [PMID: 34068811 PMCID: PMC8126238 DOI: 10.3390/s21093304] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/14/2021] [Accepted: 05/06/2021] [Indexed: 02/01/2023]
Abstract
Three-dimensional (3D) in vitro models, such as organ-on-a-chip platforms, are an emerging and effective technology that allows the replication of the function of tissues and organs, bridging the gap amid the conventional models based on planar cell cultures or animals and the complex human system. Hence, they have been increasingly used for biomedical research, such as drug discovery and personalized healthcare. A promising strategy for their fabrication is 3D printing, a layer-by-layer fabrication process that allows the construction of complex 3D structures. In contrast, 3D bioprinting, an evolving biofabrication method, focuses on the accurate deposition of hydrogel bioinks loaded with cells to construct tissue-engineered structures. The purpose of the present work is to conduct a systematic review (SR) of the published literature, according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses, providing a source of information on the evolution of organ-on-a-chip platforms obtained resorting to 3D printing and bioprinting techniques. In the literature search, PubMed, Scopus, and ScienceDirect databases were used, and two authors independently performed the search, study selection, and data extraction. The goal of this SR is to highlight the importance and advantages of using 3D printing techniques in obtaining organ-on-a-chip platforms, and also to identify potential gaps and future perspectives in this research field. Additionally, challenges in integrating sensors in organs-on-chip platforms are briefly investigated and discussed.
Collapse
Affiliation(s)
- Violeta Carvalho
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
| | - Inês Gonçalves
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
| | - Teresa Lage
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (T.L.); (R.O.R.); (G.M.)
| | - Raquel O. Rodrigues
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (T.L.); (R.O.R.); (G.M.)
| | - Graça Minas
- Center for MicroElectromechanical Systems (CMEMS-UMinho), Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (T.L.); (R.O.R.); (G.M.)
| | | | - Ana S. Moita
- IN+, Center for Innovation, Technology and Policy Research, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
- CINAMIL, Department of Exact Sciences and Engineering, Portuguese Military Academy, R. Gomes Freire 203, 1169-203 Lisboa, Portugal
| | - Takeshi Hori
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan; (T.H.); (H.K.)
| | - Hirokazu Kaji
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan; (T.H.); (H.K.)
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8579, Japan
| | - Rui A. Lima
- MEtRICs, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal;
- CEFT, Faculty of Engineering of the University of Porto (FEUP), R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
41
|
Zhao D, Lei W, Hu S. Cardiac organoid - a promising perspective of preclinical model. Stem Cell Res Ther 2021; 12:272. [PMID: 33957972 PMCID: PMC8100358 DOI: 10.1186/s13287-021-02340-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Human cardiac organoids (hCOs), three-dimensional (3D) cellular constructs similar to in vivo organ, are new-generation models. To a large extent, a hCO retains the biological characteristics and functions of cells in vivo more accurately than previous models. With the continuous development of biotechnology, the hCO model is becoming increasingly complex and mature. High-fidelity hCOs help us better explore the mysteries of human physiology and integrate phenotypes with living functions into models. Here, we discuss recent advances in the methods of constructing human cardiac organoids and introduce applications of hCOs, especially in modeling cardiovascular diseases, including myocardial infarction, heart failure, genetic cardiac diseases, and arrhythmia. In addition, we propose the prospects for and the limitations of hCOs. In conclusion, a greater understanding of hCOs will provide ways to improve hCO construction and make these models useful for future preclinical studies.
Collapse
Affiliation(s)
- Dandan Zhao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
42
|
Rae C, Amato F, Braconi C. Patient-Derived Organoids as a Model for Cancer Drug Discovery. Int J Mol Sci 2021; 22:ijms22073483. [PMID: 33801782 PMCID: PMC8038043 DOI: 10.3390/ijms22073483] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
In the search for the ideal model of tumours, the use of three-dimensional in vitro models is advancing rapidly. These are intended to mimic the in vivo properties of the tumours which affect cancer development, progression and drug sensitivity, and take into account cell–cell interactions, adhesion and invasiveness. Importantly, it is hoped that successful recapitulation of the structure and function of the tissue will predict patient response, permitting the development of personalized therapy in a timely manner applicable to the clinic. Furthermore, the use of co-culture systems will allow the role of the tumour microenvironment and tissue–tissue interactions to be taken into account and should lead to more accurate predictions of tumour development and responses to drugs. In this review, the relative merits and limitations of patient-derived organoids will be discussed compared to other in vitro and ex vivo cancer models. We will focus on their use as models for drug testing and personalized therapy and how these may be improved. Developments in technology will also be considered, including the use of microfluidics, 3D bioprinting, cryopreservation and circulating tumour cell-derived organoids. These have the potential to enhance the consistency, accessibility and availability of these models.
Collapse
Affiliation(s)
- Colin Rae
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (C.R.); (F.A.)
| | - Francesco Amato
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (C.R.); (F.A.)
| | - Chiara Braconi
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (C.R.); (F.A.)
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, UK
- Correspondence:
| |
Collapse
|
43
|
Inbody SC, Sinquefield BE, Lewis JP, Horton RE. Biomimetic microsystems for cardiovascular studies. Am J Physiol Cell Physiol 2021; 320:C850-C872. [PMID: 33760660 DOI: 10.1152/ajpcell.00026.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional tissue culture platforms have been around for several decades and have enabled key findings in the cardiovascular field. However, these platforms failed to recreate the mechanical and dynamic features found within the body. Organs-on-chips (OOCs) are cellularized microfluidic-based devices that can mimic the basic structure, function, and responses of organs. These systems have been successfully utilized in disease, development, and drug studies. OOCs are designed to recapitulate the mechanical, electrical, chemical, and structural features of the in vivo microenvironment. Here, we review cardiovascular-themed OOC studies, design considerations, and techniques used to generate these cellularized devices. Furthermore, we will highlight the advantages of OOC models over traditional cell culture vessels, discuss implementation challenges, and provide perspectives on the state of the field.
Collapse
Affiliation(s)
- Shelby C Inbody
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Bridgett E Sinquefield
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Joshua P Lewis
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Renita E Horton
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
44
|
Vargas R, Egurbide-Sifre A, Medina L. Organ-on-a-Chip systems for new drugs development. ADMET AND DMPK 2021; 9:111-141. [PMID: 35299767 PMCID: PMC8920106 DOI: 10.5599/admet.942] [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: 12/22/2020] [Revised: 03/04/2021] [Indexed: 11/18/2022] Open
Abstract
Research on alternatives to the use of animal models and cell cultures has led to the creation of organ-on-a-chip systems, in which organs and their physiological reactions to the presence of external stimuli are simulated. These systems could even replace the use of human beings as subjects for the study of drugs in clinical phases and have an impact on personalized therapies. Organ-on-a-chip technology present higher potential than traditional cell cultures for an appropriate prediction of functional impairments, appearance of adverse effects, the pharmacokinetic and toxicological profile and the efficacy of a drug. This potential is given by the possibility of placing different cell lines in a three-dimensional-arranged polymer piece and simulating and controlling specific conditions. Thus, the normal functioning of an organ, tissue, barrier, or physiological phenomenon can be simulated, as well as the interrelation between different systems. Furthermore, this alternative allows the study of physiological and pathophysiological processes. Its design combines different disciplines such as materials engineering, cell cultures, microfluidics and physiology, among others. This work presents the main considerations of OoC systems, the materials, methods and cell lines used for their design, and the conditions required for their proper functioning. Examples of applications and main challenges for the development of more robust systems are shown. This non-systematic review is intended to be a reference framework that facilitates research focused on the development of new OoC systems, as well as their use as alternatives in pharmacological, pharmacokinetic and toxicological studies.
Collapse
Affiliation(s)
- Ronny Vargas
- Industrial Pharmacy Department, Faculty of Pharmacy, University of Costa Rica 11501-2060, San José, Costa Rica
- Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-1, 08028, Barcelona, Spain
| | - Andrea Egurbide-Sifre
- Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-1, 08028, Barcelona, Spain
| | - Laura Medina
- Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-1, 08028, Barcelona, Spain
| |
Collapse
|
45
|
Chuchuy J, Rogal J, Ngo T, Stadelmann K, Antkowiak L, Achberger K, Liebau S, Schenke-Layland K, Loskill P. Integration of Electrospun Membranes into Low-Absorption Thermoplastic Organ-on-Chip. ACS Biomater Sci Eng 2021; 7:3006-3017. [DOI: 10.1021/acsbiomaterials.0c01062] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Johanna Chuchuy
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
- Department of Women’s Health, Research Institute for Women’s Health, Eberhard Karls University of Tübingen, Silcherstrasse 7/1, 72076 Tübingen, Germany
| | - Julia Rogal
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
- Department of Women’s Health, Research Institute for Women’s Health, Eberhard Karls University of Tübingen, Silcherstrasse 7/1, 72076 Tübingen, Germany
| | - Tran Ngo
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Kathrin Stadelmann
- NMI Natural and Medical Sciences Institute at the University Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Lena Antkowiak
- Institute of Neuroanatomy & Developmental Biology INDB, Eberhard Karls University of Tübingen, Österbergstraße 3, 72074 Tübingen, Germany
| | - Kevin Achberger
- Institute of Neuroanatomy & Developmental Biology INDB, Eberhard Karls University of Tübingen, Österbergstraße 3, 72074 Tübingen, Germany
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology INDB, Eberhard Karls University of Tübingen, Österbergstraße 3, 72074 Tübingen, Germany
| | - Katja Schenke-Layland
- Department of Women’s Health, Research Institute for Women’s Health, Eberhard Karls University of Tübingen, Silcherstrasse 7/1, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive South, MRL 3645, Los Angeles, California 90095, United States
| | - Peter Loskill
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
- Department of Women’s Health, Research Institute for Women’s Health, Eberhard Karls University of Tübingen, Silcherstrasse 7/1, 72076 Tübingen, Germany
| |
Collapse
|
46
|
Middelkamp HHT, Verboven AHA, De Sá Vivas AG, Schoenmaker C, Klein Gunnewiek TM, Passier R, Albers CA, 't Hoen PAC, Nadif Kasri N, van der Meer AD. Cell type-specific changes in transcriptomic profiles of endothelial cells, iPSC-derived neurons and astrocytes cultured on microfluidic chips. Sci Rep 2021; 11:2281. [PMID: 33500551 PMCID: PMC7838281 DOI: 10.1038/s41598-021-81933-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/13/2021] [Indexed: 01/30/2023] Open
Abstract
In vitro neuronal models are essential for studying neurological physiology, disease mechanisms and potential treatments. Most in vitro models lack controlled vasculature, despite its necessity in brain physiology and disease. Organ-on-chip models offer microfluidic culture systems with dedicated micro-compartments for neurons and vascular cells. Such multi-cell type organs-on-chips can emulate neurovascular unit (NVU) physiology, however there is a lack of systematic data on how individual cell types are affected by culturing on microfluidic systems versus conventional culture plates. This information can provide perspective on initial findings of studies using organs-on-chip models, and further optimizes these models in terms of cellular maturity and neurovascular physiology. Here, we analysed the transcriptomic profiles of co-cultures of human induced pluripotent stem cell (hiPSC)-derived neurons and rat astrocytes, as well as one-day monocultures of human endothelial cells, cultured on microfluidic chips. For each cell type, large gene expression changes were observed when cultured on microfluidic chips compared to conventional culture plates. Endothelial cells showed decreased cell division, neurons and astrocytes exhibited increased cell adhesion, and neurons showed increased maturity when cultured on a microfluidic chip. Our results demonstrate that culturing NVU cell types on microfluidic chips changes their gene expression profiles, presumably due to distinct surface-to-volume ratios and substrate materials. These findings inform further NVU organ-on-chip model optimization and support their future application in disease studies and drug testing.
Collapse
Affiliation(s)
- H H T Middelkamp
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands.
- BIOS/Lab on a Chip, University of Twente, Enschede, The Netherlands.
| | - A H A Verboven
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
- Centre for Molecular and Biomolecular Informatics, Radboudumc, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands.
| | - A G De Sá Vivas
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
- BIOS/Lab on a Chip, University of Twente, Enschede, The Netherlands
| | - C Schoenmaker
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - T M Klein Gunnewiek
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - R Passier
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - C A Albers
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Molecular Developmental Biology, Radboud University, Nijmegen, The Netherlands
| | - P A C 't Hoen
- Centre for Molecular and Biomolecular Informatics, Radboudumc, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands
| | - N Nadif Kasri
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Department of Cognitive Neurosciences, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - A D van der Meer
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
47
|
Agrawal G, Ramesh A, Aishwarya P, Sally J, Ravi M. Devices and techniques used to obtain and analyze three-dimensional cell cultures. Biotechnol Prog 2021; 37:e3126. [PMID: 33460298 DOI: 10.1002/btpr.3126] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022]
Abstract
Cell cultures are indispensable for both basic and applied research. Advancements in cell culture and analysis increase their utility for basic research and translational applications. A marked development in this direction is advent of three-dimensional (3D) cultures. The extent of advancement in 3D cell culture methods over the past decade has warranted referring to a single cell type being cultured as an aggregate or spheroid using simple scaffolds as "traditional." In recent years, the development of "next-generation" devices has enabled cultured cells to mimic their natural environments much better than the traditional 3D culture systems. Automated platforms like chip-based devices, magnetic- and acoustics-based assembly devices, di-electrophoresis (DEP), micro pocket cultures (MPoC), and 3D bio-printing provide a dynamic environment compared to the rather static conditions of the traditional simple scaffold-based 3D cultures. Chip-based technologies, which are centered on principles of microfluidics, are revolutionizing the ways in which cell culture and analysis can be compacted into table-top instruments. A parallel evolution in analytical devices enabled efficient assessment of various complex physiological and pathological endpoints. This is augmented by concurrent development of software enabling rapid large-scale automated data acquisition and analysis like image cytometry, elastography, optical coherence tomography, surface-enhanced Raman scattering (SERS), and biosensors. The techniques and devices utilized for the purpose of 3D cell culture and subsequent analysis depend primarily on the requirement of the study. We present here an in-depth account of the devices for obtaining and analyzing 3D cell cultures.
Collapse
Affiliation(s)
- Gatika Agrawal
- Department of Human Genetics, Faculty of Biomedical Science, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Anuradha Ramesh
- Department of Human Genetics, Faculty of Biomedical Science, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Pargaonkar Aishwarya
- Department of Human Genetics, Faculty of Biomedical Science, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Jennifer Sally
- Department of Human Genetics, Faculty of Biomedical Science, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Maddaly Ravi
- Department of Human Genetics, Faculty of Biomedical Science, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
48
|
Lu Z, Priya Rajan SA, Song Q, Zhao Y, Wan M, Aleman J, Skardal A, Bishop C, Atala A, Lu B. 3D scaffold-free microlivers with drug metabolic function generated by lineage-reprogrammed hepatocytes from human fibroblasts. Biomaterials 2021; 269:120668. [PMID: 33461059 DOI: 10.1016/j.biomaterials.2021.120668] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 01/10/2023]
Abstract
Generating microliver tissues to recapitulate hepatic function is of increasing importance in tissue engineering and drug screening. But the limited availability of primary hepatocytes and the marked loss of phenotype hinders their application. Human induced hepatocytes (hiHeps) generated by direct reprogramming can address the shortage of primary hepatocytes to make personalized drug prediction possible. Here, we simplify preparation of reprogramming reagents by expressing six transcriptional factors (HNF4A, FOXA2, FOXA3, ATF5, PROX1, and HNF1) from two lentiviral vectors, each expressing three factors. Transducing human fetal and adult fibroblasts with low vector dosage generated human induced hepatocyte-like cells (hiHeps) displaying characteristics of mature hepatocytes and capable of drug metabolism. To mimic the physiologic liver microenvironment and improve hepatocyte function, we prepared 3D scaffold-free microliver spheroids using hiHeps and human liver nonparenchymal cells through self-assembly without exogenous scaffolds. We then introduced the microliver spheroids into a two-organ microfluidic system to examine interactions between hepatocytes and tumor cells. The hiHeps-derived spheroids metabolized the prodrug capecitabine into the active metabolite 5-fluorouracil and induced toxicity in downstream tumor spheroids. Our results demonstrate that hiHeps can be used to make microliver spheroids and combined with a microfluidic system for drug evaluation. Our work could make it possible to use patient-specific hepatocyte-like cells to predict drug efficacy and side effects in various organs from the same patient.
Collapse
Affiliation(s)
- Zuyan Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Shiny Amala Priya Rajan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yu Zhao
- Jiangsu Healthy Life Innovation Medical Technology Co, Ltd., Wuxi, Jiangsu Province, China
| | - Meimei Wan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Julio Aleman
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Colin Bishop
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
49
|
Nikolakopoulou P, Rauti R, Voulgaris D, Shlomy I, Maoz BM, Herland A. Recent progress in translational engineered in vitro models of the central nervous system. Brain 2020; 143:3181-3213. [PMID: 33020798 PMCID: PMC7719033 DOI: 10.1093/brain/awaa268] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
The complexity of the human brain poses a substantial challenge for the development of models of the CNS. Current animal models lack many essential human characteristics (in addition to raising operational challenges and ethical concerns), and conventional in vitro models, in turn, are limited in their capacity to provide information regarding many functional and systemic responses. Indeed, these challenges may underlie the notoriously low success rates of CNS drug development efforts. During the past 5 years, there has been a leap in the complexity and functionality of in vitro systems of the CNS, which have the potential to overcome many of the limitations of traditional model systems. The availability of human-derived induced pluripotent stem cell technology has further increased the translational potential of these systems. Yet, the adoption of state-of-the-art in vitro platforms within the CNS research community is limited. This may be attributable to the high costs or the immaturity of the systems. Nevertheless, the costs of fabrication have decreased, and there are tremendous ongoing efforts to improve the quality of cell differentiation. Herein, we aim to raise awareness of the capabilities and accessibility of advanced in vitro CNS technologies. We provide an overview of some of the main recent developments (since 2015) in in vitro CNS models. In particular, we focus on engineered in vitro models based on cell culture systems combined with microfluidic platforms (e.g. 'organ-on-a-chip' systems). We delve into the fundamental principles underlying these systems and review several applications of these platforms for the study of the CNS in health and disease. Our discussion further addresses the challenges that hinder the implementation of advanced in vitro platforms in personalized medicine or in large-scale industrial settings, and outlines the existing differentiation protocols and industrial cell sources. We conclude by providing practical guidelines for laboratories that are considering adopting organ-on-a-chip technologies.
Collapse
Affiliation(s)
- Polyxeni Nikolakopoulou
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Rossana Rauti
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Dimitrios Voulgaris
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Iftach Shlomy
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Anna Herland
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
50
|
Richardson L, Kim S, Han A, Menon R. Modeling ascending infection with a feto-maternal interface organ-on-chip. LAB ON A CHIP 2020; 20:4486-4501. [PMID: 33112317 PMCID: PMC7815379 DOI: 10.1039/d0lc00875c] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Maternal infection (i.e., ascending infection) and the resulting host inflammatory response are risk factors associated with spontaneous preterm birth (PTB), a major pregnancy complication. However, the path of infection and its propagation from the maternal side to the fetal side have been difficult to study due to the lack of appropriate in vitro models and limitations of animal models. A better understanding of the propagation kinetics of infectious agents and development of the host inflammatory response at the feto-maternal (amniochorion-decidua, respectively) interface (FMi) is critical in curtailing host inflammatory responses that can lead to PTB. To model ascending infection and determine inflammatory responses at the FMi, we developed a microfluidic organ-on-chip (OOC) device containing primary cells from the FMi (decidua, chorion, and amnion [mesenchyme and epithelium]) and collagen matrix harvested from primary tissue. The FMi-OOC is composed of four concentric circular cell/collagen chambers designed to mimic the thickness and cell density of the FMi in vivo. Each layer is connected by arrays of microchannels filled with type IV collagen to recreate the basement membrane of the amniochorion. Cellular characteristics (viability, morphology, production of nascent collagen, cellular transitions, and migration) in the OOC were similar to those seen in utero, validating the physiological relevance and utility of the developed FMi-OOC. The ascending infection model of the FMi-OOC, triggered by exposing the maternal (decidua) side of the OOC to lipopolysaccharide (LPS, 100 ng mL-1), shows that LPS propagated through the chorion, amnion mesenchyme, and reached the fetal amnion within 72 h. LPS induced time-dependent and cell-type-specific pro-inflammatory cytokine production (24 h decidua: IL-6, 48 h chorion: GM-CSF and IL-6, and 72 h amnion mesenchyme and epithelium: GM-CSF and IL-6). Collectively, this OOC model and study successfully modeled ascending infection, its propagation, and distinct inflammatory response at the FMi indicative of pathologic pathways of PTB. This OOC model provides a novel platform to study physiological and pathological cell status at the FMi, and is expected to have broad utility in the field of obstetrics.
Collapse
Affiliation(s)
- Lauren Richardson
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA
| |
Collapse
|