1
|
Geng Q, Xu Y, Hu Y, Wang L, Wang Y, Fan Z, Kong D. Progress in the Application of Organoids-On-A-Chip in Diseases. Organogenesis 2024; 20:2386727. [PMID: 39126669 PMCID: PMC11318694 DOI: 10.1080/15476278.2024.2386727] [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: 03/05/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
With the rapid development of the field of life sciences, traditional 2D cell culture and animal models have long been unable to meet the urgent needs of modern biomedical research and new drug development. Establishing a new generation of experimental models and research models is of great significance for deeply understanding human health and disease processes, and developing effective treatment measures. As is well known, long research and development cycles, high risks, and high costs are the "three mountains" facing the development of new drugs today. Organoids and organ-on-chips technology can highly simulate and reproduce the human physiological environment and complex reactions in vitro, greatly improving the accuracy of drug clinical efficacy prediction, reducing drug development costs, and avoiding the defects of drug testing animal models. Therefore, organ-on-chips have enormous potential in medical diagnosis and treatment.
Collapse
Affiliation(s)
- Qiao Geng
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanyan Xu
- Department of Anoenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Hu
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Wang
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Wang
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhimin Fan
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Pun S, Prakash A, Demaree D, Krummel DP, Sciumè G, Sengupta S, Barrile R. Rapid Biofabrication of an Advanced Microphysiological System Mimicking Phenotypical Heterogeneity and Drug Resistance in Glioblastoma. Adv Healthc Mater 2024; 13:e2401876. [PMID: 39101329 PMCID: PMC11616263 DOI: 10.1002/adhm.202401876] [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: 05/20/2024] [Revised: 07/10/2024] [Indexed: 08/06/2024]
Abstract
Microphysiological systems (MPSs) reconstitute tissue interfaces and organ functions, presenting a promising alternative to animal models in drug development. However, traditional materials like polydimethylsiloxane (PDMS) often interfere by absorbing hydrophobic molecules, affecting drug testing accuracy. Additive manufacturing, including 3D bioprinting, offers viable solutions. GlioFlow3D, a novel microfluidic platform combining extrusion bioprinting and stereolithography (SLA) is introduced. GlioFlow3D integrates primary human cells and glioblastoma (GBM) lines in hydrogel-based microchannels mimicking vasculature, within an SLA resin framework using cost-effective materials. The study introduces a robust protocol to mitigate SLA resin cytotoxicity. Compared to PDMS, GlioFlow3D demonstrated lower small molecule absorption, which is relevant for accurate testing of small molecules like Temozolomide (TMZ). Computational modeling is used to optimize a pumpless setup simulating interstitial fluid flow dynamics in tissues. Co-culturing GBM with brain endothelial cells in GlioFlow3D showed enhanced CD133 expression and TMZ resistance near vascular interfaces, highlighting spatial drug resistance mechanisms. This PDMS-free platform promises advanced drug testing, improving preclinical research and personalized therapy by elucidating complex GBM drug resistance mechanisms influenced by the tissue microenvironment.
Collapse
Affiliation(s)
- Sirjana Pun
- Department of Biomedical EngineeringUniversity of CincinnatiCincinnatiOH45221USA
| | - Anusha Prakash
- Department of Biomedical EngineeringUniversity of CincinnatiCincinnatiOH45221USA
- AbbvieWorcesterMassachusetts01605USA
| | - Dalee Demaree
- Department of Biomedical EngineeringUniversity of CincinnatiCincinnatiOH45221USA
- Thermo Fisher ScientificWalthamMassachusetts02451USA
| | - Daniel Pomeranz Krummel
- Department of NeurologyUniversity of CincinnatiCincinnatiOH45219USA
- Department of NeurosurgeryUniversity of North CarolinaChapel HillNC27599USA
| | - Giuseppe Sciumè
- Institute of Mechanics and Engineering‐12 MUniversity of BordeauxBordeaux33607France
| | - Soma Sengupta
- Department of NeurologyUniversity of CincinnatiCincinnatiOH45219USA
- Department of NeurosurgeryUniversity of North CarolinaChapel HillNC27599USA
- Department of NeurologyUniversity of North CarolinaChapel HillNC27599‐7025USA
- Lineberger Comprehensive Cancer CenterUniversity of North CarolinaChapel HillNC27599‐7295USA
| | - Riccardo Barrile
- Department of Biomedical EngineeringUniversity of CincinnatiCincinnatiOH45221USA
- Center for Stem Cells and Organoid Medicine (CuSTOM)Cincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
| |
Collapse
|
3
|
Yilmaz EG, Hacıosmanoğlu N, Jordi SBU, Yilmaz B, Inci F. Revolutionizing IBD research with on-chip models of disease modeling and drug screening. Trends Biotechnol 2024:S0167-7799(24)00284-1. [PMID: 39523166 DOI: 10.1016/j.tibtech.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) comprises chronic inflammatory conditions with complex mechanisms and diverse manifestations, posing significant clinical challenges. Traditional animal models and ethical concerns in human studies necessitate innovative approaches. This review provides an overview of human intestinal architecture in health and inflammation, emphasizing the role of microfluidics and on-chip technologies in IBD research. These technologies allow precise manipulation of cellular and microbial interactions in a physiologically relevant context, simulating the intestinal ecosystem microscopically. By integrating cellular components and replicating 3D tissue architecture, they offer promising models for studying host-microbe interactions, wound healing, and therapeutic approaches. Continuous refinement of these technologies promises to advance IBD understanding and therapy development, inspiring further innovation and cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Eylul Gulsen Yilmaz
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Nedim Hacıosmanoğlu
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Sebastian Bruno Ulrich Jordi
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland.
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey.
| |
Collapse
|
4
|
Ebrahimi A, Ak G, Özel C, İzgördü H, Ghorbanpoor H, Hassan S, Avci H, Metintaş M. Clinical Perspectives and Novel Preclinical Models of Malignant Pleural Mesothelioma: A Critical Review. ACS Pharmacol Transl Sci 2024; 7:3299-3333. [PMID: 39539262 PMCID: PMC11555512 DOI: 10.1021/acsptsci.4c00324] [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: 05/31/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Pleural mesothelioma (PM), a rare malignant tumor explicitly associated with asbestos and erionite exposures, has become a global health problem due to limited treatment options and a poor prognosis, in which the median life expectancy varies depending on the method of treatment. However, the importance of early diagnosis is emphasized, and the practical methods have not matured yet. This study provides a critical overview of PM, addressing various aspects like epidemiology, etiology, diagnosis, treatment options, and the potential use of advanced technologies like microfluidic chip-based models for research and diagnosis. It initially begins with fundamentals of clinical aspects and then discusses the identification of disease-specific biomarkers in patients' serum or plasma samples, which could potentially be used for early diagnosis. A detailed investigation of the sophisticated preclinical models is highlighted. Recent three-dimensional (3D) model accomplishments, including microarchitecture modeling by transwell coculture, spheroids, organoids, 3D bioprinting constructs, and ex vivo tumor slices, are discussed comprehensively. On-chip models that imitate physiological processes, such as detection chips and therapeutic screening chips, are assessed as potential techniques. The review concludes with a critical and constructive discussion of the growing interest in the topic and its limitations and suggestions.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Güntülü Ak
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Ceren Özel
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Hüseyin İzgördü
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Hamed Ghorbanpoor
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Biomedical Engineering, Eskişehir
Osmangazi University, Eskişehir 26040, Turkey
| | - Shabir Hassan
- Department
of Biological Sciences, Khalifa University
of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Huseyin Avci
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Muzaffer Metintaş
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| |
Collapse
|
5
|
Michielon E, Boninsegna M, Waaijman T, Fassini D, Spiekstra SW, Cramer J, Gaudriault P, Kodolányi J, de Gruijl TD, Homs-Corbera A, Gibbs S. Environmentally Controlled Microfluidic System Enabling Immune Cell Flow and Activation in an Endothelialised Skin-On-Chip. Adv Healthc Mater 2024; 13:e2400750. [PMID: 39370595 PMCID: PMC11582514 DOI: 10.1002/adhm.202400750] [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: 02/27/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Integration of reconstructed human skin (RhS) into organ-on-chip (OoC) platforms addresses current limitations imposed by static culturing. This innovation, however, is not without challenges. Microfluidic devices, while powerful, often encounter usability, robustness, and gas bubble issues that hinder large-scale high-throughput setups. This study aims to develop a novel re-usable multi-well microfluidic adaptor (MMA) with the objective to provide a flexible tool for biologists implementing complex 3D biological models (e.g., skin) while enabling simultaneous user control over temperature, medium flow, oxigen (O2), nitrogen (N2), and carbon dioxide (CO2) without the need for an incubator. The presented MMA device is designed to be compatible with standard, commercially available 6-well multi-well plates (6MWPs) and 12-well transwells. This MMA-6MWP setup is employed to generate a skin-on-chip (SoC). RhS viability is maintained under flow for three days and their morphology closely resembles that of native human skin. A proof-of-concept study demonstrates the system's potential in toxicology applications by combining endothelialised RhS with flowing immune cells. This dynamic setting activates the monocyte-like MUTZ-3 cells (CD83 and CD86 upregulation) upon topical exposure of RhS to a sensitizer, revealing the MMA-6MWP's unique capabilities compared to static culturing, where such activation is absent.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
| | - Matteo Boninsegna
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
- Department of Physics, Bielefeld University, Universitätsstr 25, 33615, Bielefeld, Germany
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Dario Fassini
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jeremy Cramer
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Pierre Gaudriault
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - János Kodolányi
- Department of Dental Material Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Antoni Homs-Corbera
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| |
Collapse
|
6
|
Li J, Han S, Yu F, Li T, Liao B, Liu F. Mapping the landscape of PSC-CM research through bibliometric analysis. Front Cardiovasc Med 2024; 11:1435874. [PMID: 39450232 PMCID: PMC11499114 DOI: 10.3389/fcvm.2024.1435874] [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: 05/21/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Objectives The discovery of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) has not only deepened our understanding of the pathogenesis and progression of heart disease, but also advanced the development of engineered cardiac tissues, cardiac regenerative therapy, drug discovery and the cardiotoxicity assessment of drugs. This study aims to visualize the developmental trajectory of PSC-CM research over the past 18 years to identify the emerging research frontiers and challenges. Methods The literature on PSC-CMs from 2007 to 2024 was retrieved from the Web of Science and PubMed databases. Bibliometrix, VOSviewer and CiteSpace software were used for statistical analysis and visualization of scientific literature. Previous clinical trials were summarized using data from the ClinicalTrials.gov database. Results A total of 29,660 authors from 81 countries and regions published 6,406 papers on PSC-CMs over the past 18 years. The annual output of PSC-CM research experienced a general upward trend from 2007 to 2021, reaching its peak in 2021, followed by a notable decline in 2022 and 2023. The United States has emerged as the most influential nation in this field, with Stanford University being the most prolific institution and Joseph C. Wu standing out as the most productive and highly cited scholar. Circulation Research, Circulation, and Nature have been identified as the most co-cited journals. Organ-on-a-chip, 3D bio-printing, cardiac microtissue, extracellular vesicle, inflammation, energy metabolism, atrial fibrillation, personalized medicine etc., with a longer burst period, and maturation of PSC-CMs, with the highest burst strength of 27.19, are the major research focuses for rigorous investigation in recent years. Cardiac organoid is emerging as a promising key research frontier. While the clinical trials of stem-cell-mediated treatment for heart diseases shows promise, significant challenges remain. Further research is imperative to optimize protocols, enhance cell delivery methods, and establish standardized practices to improve clinical outcomes. Conclusions In conclusion, several major research hotspots, including engineered cardiac tissue and maturation, exosome-based regenerative therapy, inflammation response, energy metabolism, atrial fibrillation, and personalized medicine etc. will continue to attract substantial interest from investigators worldwide. Cardiac organoids to in vitro recapitulate the intricate human heart is emerging as a promising key research frontier. Significant challenges persist in the clinical trials of stem-cell-mediated therapies for heart diseases.
Collapse
Affiliation(s)
- Jun Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Shangting Han
- Department of Organ Transplantation, Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Bin Liao
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Feng Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular Remodeling and Dysfunction Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| |
Collapse
|
7
|
Mihailovich M, Soković Bajić S, Dinić M, Đokić J, Živković M, Radojević D, Golić N. Cutting-Edge iPSC-Based Approaches in Studying Host-Microbe Interactions in Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:10156. [PMID: 39337640 PMCID: PMC11432053 DOI: 10.3390/ijms251810156] [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: 08/21/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Gut microbiota (GM), together with its metabolites (such as SCFA, tryptophan, dopamine, GABA, etc.), plays an important role in the functioning of the central nervous system. Various neurological and psychiatric disorders are associated with changes in the composition of GM and their metabolites, which puts them in the foreground as a potential adjuvant therapy. However, the molecular mechanisms behind this relationship are not clear enough. Therefore, before considering beneficial microbes and/or their metabolites as potential therapeutics for brain disorders, the mechanisms underlying microbiota-host interactions must be identified and characterized in detail. In this review, we summarize the current knowledge of GM alterations observed in prevalent neurological and psychiatric disorders, multiple sclerosis, major depressive disorder, Alzheimer's disease, and autism spectrum disorders, together with experimental evidence of their potential to improve patients' quality of life. We further discuss the main obstacles in the study of GM-host interactions and describe the state-of-the-art solution and trends in this field, namely "culturomics" which enables the culture and identification of novel bacteria that inhabit the human gut, and models of the gut and blood-brain barrier as well as the gut-brain axis based on induced pluripotent stem cells (iPSCs) and iPSC derivatives, thus pursuing a personalized medicine agenda for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Marija Mihailovich
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
- Human Technopole, Palazzo Italia, Viale Rita Levi-Montalcini, 1, 20157 Milan, Italy
| | - Svetlana Soković Bajić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
| | - Miroslav Dinić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
| | - Milica Živković
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
| | - Dušan Radojević
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
| | - Nataša Golić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.B.); (M.D.); (J.Đ.); (M.Ž.); (D.R.)
| |
Collapse
|
8
|
Papp D, Korcsmaros T, Hautefort I. Revolutionizing immune research with organoid-based co-culture and chip systems. Clin Exp Immunol 2024; 218:40-54. [PMID: 38280212 PMCID: PMC11404127 DOI: 10.1093/cei/uxae004] [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: 06/27/2023] [Revised: 12/05/2023] [Accepted: 01/24/2024] [Indexed: 01/29/2024] Open
Abstract
The intertwined interactions various immune cells have with epithelial cells in our body require sophisticated experimental approaches to be studied. Due to the limitations of immortalized cell lines and animal models, there is an increasing demand for human in vitro model systems to investigate the microenvironment of immune cells in normal and in pathological conditions. Organoids, which are self-renewing, 3D cellular structures that are derived from stem cells, have started to provide gap-filling tissue modelling solutions. In this review, we first demonstrate with some of the available examples how organoid-based immune cell co-culture experiments can advance disease modelling of cancer, inflammatory bowel disease, and tissue regeneration. Then, we argue that to achieve both complexity and scale, organ-on-chip models combined with cutting-edge microfluidics-based technologies can provide more precise manipulation and readouts. Finally, we discuss how genome editing techniques and the use of patient-derived organoids and immune cells can improve disease modelling and facilitate precision medicine. To achieve maximum impact and efficiency, these efforts should be supported by novel infrastructures such as organoid biobanks, organoid facilities, as well as drug screening and host-microbe interaction testing platforms. All these together or in combination can allow researchers to shed more detailed, and often patient-specific, light on the crosstalk between immune cells and epithelial cells in health and disease.
Collapse
Affiliation(s)
- Diana Papp
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- NIHR Imperial BRC Organoid Facility, Imperial College London, London, UK
| | - Tamas Korcsmaros
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- NIHR Imperial BRC Organoid Facility, Imperial College London, London, UK
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Isabelle Hautefort
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- NIHR Imperial BRC Organoid Facility, Imperial College London, London, UK
- Food, Microbiome and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
| |
Collapse
|
9
|
Filiz Y, Esposito A, De Maria C, Vozzi G, Yesil-Celiktas O. A comprehensive review on organ-on-chips as powerful preclinical models to study tissue barriers. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 6:042001. [PMID: 39655848 DOI: 10.1088/2516-1091/ad776c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 09/04/2024] [Indexed: 12/18/2024]
Abstract
In the preclinical stage of drug development, 2D and 3D cell cultures under static conditions followed by animal models are utilized. However, these models are insufficient to recapitulate the complexity of human physiology. With the developing organ-on-chip (OoC) technology in recent years, human physiology and pathophysiology can be modeled better than traditional models. In this review, the need for OoC platforms is discussed and evaluated from both biological and engineering perspectives. The cellular and extracellular matrix components are discussed from a biological perspective, whereas the technical aspects such as the intricate working principles of these systems, the pivotal role played by flow dynamics and sensor integration within OoCs are elucidated from an engineering perspective. Combining these two perspectives, bioengineering applications are critically discussed with a focus on tissue barriers such as blood-brain barrier, ocular barrier, nasal barrier, pulmonary barrier and gastrointestinal barrier, featuring recent examples from the literature. Furthermore, this review offers insights into the practical utility of OoC platforms for modeling tissue barriers, showcasing their potential and drawbacks while providing future projections for innovative technologies.
Collapse
Affiliation(s)
- Yagmur Filiz
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 8500 Kortrijk, Belgium
| | - Alessio Esposito
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- EgeSAM-Ege University Translational Pulmonary Research Center, Bornova, Izmir, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| |
Collapse
|
10
|
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
|
11
|
Hu W, Bei HP, Jiang H, Wu D, Yu X, Zhou X, Sun Q, Lu Q, Du Q, Wang L, Luo Z, Wu G, Zhao X, Wang S. DLM-GelMA/tumor slice sandwich structured tumor on a chip for drug efficacy testing. LAB ON A CHIP 2024; 24:3718-3727. [PMID: 38953554 DOI: 10.1039/d4lc00278d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The in vitro recapitulation of tumor microenvironment is of great interest to preclinical screening of drugs. Compared with culture of cell lines, tumor organ slices can better preserve the complex tumor architecture and phenotypic activity of native cells, but are limited by their exposure to fluid shear and gradual degradation under perfusion culture. Here, we established a decellularized liver matrix (DLM)-GelMA "sandwich" structure and a perfusion-based microfluidic platform to support long-term culture of tumor slices with excellent structural integrity and cell viability over 7 days. The DLM-GelMA was able to secrete cytokines and growth factors while providing shear protection to the tumor slice via the sandwich structure, leading to the preservation of the tumor microenvironment where immune cells (CD3, CD8, CD68), tumor-associated fibroblasts (α-SMA), and extracellular matrix components (collagen I, fibronectin) were well maintained. Furthermore, this chip presented anti-tumor efficacy at cisplatin (20 μM) on tumor patients, demonstrating our platform's efficacy to design patient-specific treatment regimens. Taken together, the successful development of this DLM-GelMA sandwich structure on the chip could faithfully reflect the tumor microenvironment and immune response, accelerating the screening process of drug molecules and providing insights for practical medicine.
Collapse
Affiliation(s)
- Wenqi Hu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Ho-Pan Bei
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
| | - Hongwei Jiang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Di Wu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Xiaorui Yu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Xintong Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
| | - Qiuwan Sun
- Sichuan Diya BioTechnology Group Company, Chengdu, 641400, China
| | - Qinrui Lu
- Sichuan Diya BioTechnology Group Company, Chengdu, 641400, China
| | - Qijun Du
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Liangwen Wang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhi Luo
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Guohua Wu
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
- Research Institute for Intelligent Wearable Systems, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, 999077, China
| | - Shuqi Wang
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| |
Collapse
|
12
|
Jadalannagari S, Ewart L. Beyond the hype and toward application: liver complex in vitro models in preclinical drug safety. Expert Opin Drug Metab Toxicol 2024; 20:607-619. [PMID: 38465923 DOI: 10.1080/17425255.2024.2328794] [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: 01/15/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Drug induced Liver-Injury (DILI) is a leading cause of drug attrition and complex in vitro models (CIVMs), including three dimensional (3D) spheroids, 3D bio printed tissues and flow-based systems, could improve preclinical prediction. Although CIVMs have demonstrated good sensitivity and specificity in DILI detection their adoption remains limited. AREAS COVERED This article describes DILI, the challenges with its prediction and the current strategies and models that are being used. It reviews data from industry-FDA collaborations and strategic partnerships and finishes with an outlook of CIVMs in preclinical toxicity testing. Literature searches were performed using PubMed and Google Scholar while product information was collected from manufacturer websites. EXPERT OPINION Liver CIVMs are promising models for predicting DILI although, a decade after their introduction, routine use by the pharmaceutical industry is limited. To accelerate their adoption, several industry-regulator-developer partnerships or consortia have been established to guide the development and qualification. Beyond this, liver CIVMs should continue evolving to capture greater immunological mimicry while partnering with computational approaches to deliver systems that change the paradigm of predicting DILI.
Collapse
Affiliation(s)
| | - Lorna Ewart
- Department of Bioinnovations, Emulate Inc, Boston, MA, USA
| |
Collapse
|
13
|
Katirai A. The Environmental Costs of Artificial Intelligence for Healthcare. Asian Bioeth Rev 2024; 16:527-538. [PMID: 39022383 PMCID: PMC11250743 DOI: 10.1007/s41649-024-00295-4] [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: 01/26/2024] [Revised: 03/09/2024] [Accepted: 03/26/2024] [Indexed: 07/20/2024] Open
Abstract
Healthcare has emerged as a key setting where expectations are rising for the potential benefits of artificial intelligence (AI), encompassing a range of technologies of varying utility and benefit. This paper argues that, even as the development of AI for healthcare has been pushed forward by a range of public and private actors, insufficient attention has been paid to a key contradiction at the center of AI for healthcare: that its pursuit to improve health is necessarily accompanied by environmental costs which pose risks to human and environmental health-costs which are not necessarily directly borne by those benefiting from the technologies. This perspective paper begins by examining the purported promise of AI in healthcare, contrasting this with the environmental costs which arise across the AI lifecycle, to highlight this contradiction inherent in the pursuit of AI. Its advancement-including in healthcare-is often described through deterministic language that presents it as inevitable. Yet, this paper argues that there is need for recognition of the environmental harm which this pursuit can lead to. Given recent initiatives to incorporate stakeholder involvement into decision-making around AI, the paper closes with a call for an expanded conception of stakeholders in AI for healthcare, to include consideration of those who may be indirectly affected by its development and deployment.
Collapse
Affiliation(s)
- Amelia Katirai
- Research Center on Ethical, Legal, and Social Issues, Osaka University, Osaka, Japan
| |
Collapse
|
14
|
Kumar D, Nadda R, Repaka R. Advances and challenges in organ-on-chip technology: toward mimicking human physiology and disease in vitro. Med Biol Eng Comput 2024; 62:1925-1957. [PMID: 38436835 DOI: 10.1007/s11517-024-03062-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
Organs-on-chips have been tissues or three-dimensional (3D) mini-organs that comprise numerous cell types and have been produced on microfluidic chips to imitate the complicated structures and interactions of diverse cell types and organs under controlled circumstances. Several morphological and physiological distinctions exist between traditional 2D cultures, animal models, and the growing popular 3D cultures. On the other hand, animal models might not accurately simulate human toxicity because of physiological variations and interspecies metabolic capability. The on-chip technique allows for observing and understanding the process and alterations occurring in metastases. The present study aimed to briefly overview single and multi-organ-on-chip techniques. The current study addresses each platform's essential benefits and characteristics and highlights recent developments in developing and utilizing technologies for single and multi-organs-on-chips. The study also discusses the drawbacks and constraints associated with these models, which include the requirement for standardized procedures and the difficulties of adding immune cells and other intricate biological elements. Finally, a comprehensive review demonstrated that the organs-on-chips approach has a potential way of investigating organ function and disease. The advancements in single and multi-organ-on-chip structures can potentially increase drug discovery and minimize dependency on animal models, resulting in improved therapies for human diseases.
Collapse
Affiliation(s)
- Dhiraj Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
| | - Rahul Nadda
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India.
| | - Ramjee Repaka
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, 140001, India
| |
Collapse
|
15
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [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: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
16
|
Mou X, Shah J, Roye Y, Du C, Musah S. An ultrathin membrane mediates tissue-specific morphogenesis and barrier function in a human kidney chip. SCIENCE ADVANCES 2024; 10:eadn2689. [PMID: 38838141 PMCID: PMC11152122 DOI: 10.1126/sciadv.adn2689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/30/2024] [Indexed: 06/07/2024]
Abstract
Organ-on-chip (OOC) systems are revolutionizing tissue engineering by providing dynamic models of tissue structure, organ-level function, and disease phenotypes using human cells. However, nonbiological components of OOC devices often limit the recapitulation of in vivo-like tissue-tissue cross-talk and morphogenesis. Here, we engineered a kidney glomerulus-on-a-chip that recapitulates glomerular morphogenesis and barrier function using a biomimetic ultrathin membrane and human-induced pluripotent stem cells. The resulting chip comprised a proximate epithelial-endothelial tissue interface, which reconstituted the selective molecular filtration function of healthy and diseased kidneys. In addition, fenestrated endothelium was successfully induced from human pluripotent stem cells in an OOC device, through in vivo-like paracrine signaling across the ultrathin membrane. Thus, this device provides a dynamic tissue engineering platform for modeling human kidney-specific morphogenesis and function, enabling mechanistic studies of stem cell differentiation, organ physiology, and pathophysiology.
Collapse
Affiliation(s)
- Xingrui Mou
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Jessica Shah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Carolyn Du
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27710, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Affiliate Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
17
|
Shariff S, Kantawala B, Xochitun Gopar Franco W, Dejene Ayele N, Munyangaju I, Esam Alzain F, Nazir A, Wojtara M, Uwishema O. Tailoring epilepsy treatment: personalized micro-physiological systems illuminate individual drug responses. Ann Med Surg (Lond) 2024; 86:3557-3567. [PMID: 38846814 PMCID: PMC11152789 DOI: 10.1097/ms9.0000000000002078] [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: 11/09/2023] [Accepted: 04/09/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Approximately 50 million people worldwide have epilepsy, with many not achieving seizure freedom. Organ-on-chip technology, which mimics organ-level physiology, could revolutionize drug development for epilepsy by replacing animal models in preclinical studies. The authors' goal is to determine if customized micro-physiological systems can lead to tailored drug treatments for epileptic patients. Materials and methods A comprehensive literature search was conducted utilizing various databases, including PubMed, Ebscohost, Medline, and the National Library of Medicine, using a predetermined search strategy. The authors focused on articles that addressed the role of personalized micro-physiological systems in individual drug responses and articles that discussed different types of epilepsy, diagnosis, and current treatment options. Additionally, articles that explored the components and design considerations of micro-physiological systems were reviewed to identify challenges and opportunities in drug development for challenging epilepsy cases. Results The micro-physiological system offers a more accurate and cost-effective alternative to traditional models for assessing drug effects, toxicities, and disease mechanisms. Nevertheless, designing patient-specific models presents critical considerations, including the integration of analytical biosensors and patient-derived cells, while addressing regulatory, material, and biological complexities. Material selection, standardization, integration of vascular systems, cost efficiency, real-time monitoring, and ethical considerations are also crucial to the successful use of this technology in drug development. Conclusion The future of organ-on-chip technology holds great promise, with the potential to integrate artificial intelligence and machine learning for personalized treatment of epileptic patients.
Collapse
Affiliation(s)
- Sanobar Shariff
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - Burhan Kantawala
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - William Xochitun Gopar Franco
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- University of Guadalajara, Guadalajara, Mexico
| | - Nitsuh Dejene Ayele
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Internal Medicine, Faculty of Medicine, Wolkite University, Wolkite, Ethiopia
| | - Isabelle Munyangaju
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Medicine and General Surgery, Sudan University Of Science and Technology, Khartoum, Sudan
| | - Fatima Esam Alzain
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Medicine and General Surgery, Sudan University Of Science and Technology, Khartoum, Sudan
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Madga Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Clinton Global Initiative University, New York, NY
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
18
|
Kim S, Lam PY, Jayaraman A, Han A. Uniform sized cancer spheroids production using hydrogel-based droplet microfluidics: a review. Biomed Microdevices 2024; 26:26. [PMID: 38806765 PMCID: PMC11241584 DOI: 10.1007/s10544-024-00712-3] [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] [Accepted: 05/16/2024] [Indexed: 05/30/2024]
Abstract
Three-dimensional (3D) cell culture models have been extensively utilized in various mechanistic studies as well as for drug development studies as superior in vitro platforms than conventional two-dimensional (2D) cell culture models. This is especially the case in cancer biology, where 3D cancer models, such as spheroids or organoids, have been utilized extensively to understand the mechanisms of cancer development. Recently, many sophisticated 3D models such as organ-on-a-chip models are emerging as advanced in vitro models that can more accurately mimic the in vivo tissue functions. Despite such advancements, spheroids are still considered as a powerful 3D cancer model due to the relatively simple structure and compatibility with existing laboratory instruments, and also can provide orders of magnitude higher throughput than complex in vitro models, an extremely important aspects for drug development. However, creating well-defined spheroids remain challenging, both in terms of throughputs in generation as well as reproducibility in size and shape that can make it challenging for drug testing applications. In the past decades, droplet microfluidics utilizing hydrogels have been highlighted due to their potentials. Importantly, core-shell structured gel droplets can avoid spheroid-to-spheroid adhesion that can cause large variations in assays while also enabling long-term cultivation of spheroids with higher uniformity by protecting the core organoid area from external environment while the outer porous gel layer still allows nutrient exchange. Hence, core-shell gel droplet-based spheroid formation can improve the predictivity and reproducibility of drug screening assays. This review paper will focus on droplet microfluidics-based technologies for cancer spheroid production using various gel materials and structures. In addition, we will discuss emerging technologies that have the potential to advance the production of spheroids, prospects of such technologies, and remaining challenges.
Collapse
Affiliation(s)
- Sungjin Kim
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Po Yi Lam
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [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: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
20
|
De Lazzari G, Opattova A, Arena S. Novel frontiers in urogenital cancers: from molecular bases to preclinical models to tailor personalized treatments in ovarian and prostate cancer patients. J Exp Clin Cancer Res 2024; 43:146. [PMID: 38750579 PMCID: PMC11094891 DOI: 10.1186/s13046-024-03065-0] [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: 01/31/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
Over the last few decades, the incidence of urogenital cancers has exhibited diverse trends influenced by screening programs and geographical variations. Among women, there has been a consistent or even increased occurrence of endometrial and ovarian cancers; conversely, prostate cancer remains one of the most diagnosed malignancies, with a rise in reported cases, partly due to enhanced and improved screening efforts.Simultaneously, the landscape of cancer therapeutics has undergone a remarkable evolution, encompassing the introduction of targeted therapies and significant advancements in traditional chemotherapy. Modern targeted treatments aim to selectively address the molecular aberrations driving cancer, minimizing adverse effects on normal cells. However, traditional chemotherapy retains its crucial role, offering a broad-spectrum approach that, despite its wider range of side effects, remains indispensable in the treatment of various cancers, often working synergistically with targeted therapies to enhance overall efficacy.For urogenital cancers, especially ovarian and prostate cancers, DNA damage response inhibitors, such as PARP inhibitors, have emerged as promising therapeutic avenues. In BRCA-mutated ovarian cancer, PARP inhibitors like olaparib and niraparib have demonstrated efficacy, leading to their approval for specific indications. Similarly, patients with DNA damage response mutations have shown sensitivity to these agents in prostate cancer, heralding a new frontier in disease management. Furthermore, the progression of ovarian and prostate cancer is intricately linked to hormonal regulation. Ovarian cancer development has also been associated with prolonged exposure to estrogen, while testosterone and its metabolite dihydrotestosterone, can fuel the growth of prostate cancer cells. Thus, understanding the interplay between hormones, DNA damage and repair mechanisms can hold promise for exploring novel targeted therapies for ovarian and prostate tumors.In addition, it is of primary importance the use of preclinical models that mirror as close as possible the biological and genetic features of patients' tumors in order to effectively translate novel therapeutic findings "from the bench to the bedside".In summary, the complex landscape of urogenital cancers underscores the need for innovative approaches. Targeted therapy tailored to DNA repair mechanisms and hormone regulation might offer promising avenues for improving the management and outcomes for patients affected by ovarian and prostate cancers.
Collapse
Affiliation(s)
- Giada De Lazzari
- Candiolo Cancer Institute, FPO - IRCCS, Laboratory of Translational Cancer Genetics, Strada Provinciale 142, Km 3.95, Candiolo, TO, ZIP 10060, Italy
| | - Alena Opattova
- Candiolo Cancer Institute, FPO - IRCCS, Laboratory of Translational Cancer Genetics, Strada Provinciale 142, Km 3.95, Candiolo, TO, ZIP 10060, Italy
| | - Sabrina Arena
- Candiolo Cancer Institute, FPO - IRCCS, Laboratory of Translational Cancer Genetics, Strada Provinciale 142, Km 3.95, Candiolo, TO, ZIP 10060, Italy.
- Department of Oncology, University of Torino, Strada Provinciale 142, Km 3.95, Candiolo, TO, ZIP 10060, Italy.
| |
Collapse
|
21
|
Hsu YH, Yang WC, Chen YT, Lin CY, Yang CF, Liu WW, Shivani S, Li PC. Spatially controlled diffusion range of tumor-associated angiogenic factors to develop a tumor model using a microfluidic resistive circuit. LAB ON A CHIP 2024; 24:2644-2657. [PMID: 38576341 DOI: 10.1039/d3lc00891f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Developing a tumor model with vessels has been a challenge in microfluidics. This difficulty is because cancer cells can overgrow in a co-culture system. The up-regulation of anti-angiogenic factors during the initial tumor development can hinder neovascularization. The standard method is to develop a quiescent vessel network before loading a tumor construct in an adjacent chamber, which simulates the interaction between a tumor and its surrounding vessels. Here, we present a new method that allows a vessel network and a tumor to develop simultaneously in two linked chambers. The physiological environment of these two chambers is controlled by a microfluidic resistive circuit using two symmetric long microchannels. Applying the resistive circuit, a diffusion-dominated environment with a small 2-D pressure gradient is created across the two chambers with velocity <10.9 nm s-1 and Péclet number <6.3 × 10-5. This 2-D pressure gradient creates a V-shaped velocity clamp to confine the tumor-associated angiogenic factors at pores between the two chambers, and it has two functions. At the early stage, vasculogenesis is stimulated to grow a vessel network in the vessel chamber with minimal influence from the tumor that is still developed in the adjacent chamber. At the post-tumor-development stage, the induced steep concentration gradient at pores mimics vessel-tumor interactions to stimulate angiogenesis to grow vessels toward the tumor. Applying this method, we demonstrate that vasculogenic vessels can grow first, followed by stimulating angiogenesis. Angiogenic vessels can grow into stroma tissue up to 1.3 mm long, and vessels can also grow into or wrap around a 625 μm tumor spheroid or a tumor tissue developed from a cell suspension. In summary, our study suggests that the interactions between a developing vasculature and a growing tumor must be controlled differently throughout the tissue development process, including at the early stage when vessels are still forming and at the later stage when the tumor needs to interact with the vessels.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
- Graduate School of Advanced Technology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Wen-Chih Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Yi-Ting Chen
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Che-Yu Lin
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Chiou-Fong Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Wei-Wen Liu
- Graduate Institute of Oral Biology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Subhashree Shivani
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| | - Pai-Chi Li
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
22
|
Dasgupta I, Rangineni DP, Abdelsaid H, Ma Y, Bhushan A. Tiny Organs, Big Impact: How Microfluidic Organ-on-Chip Technology Is Revolutionizing Mucosal Tissues and Vasculature. Bioengineering (Basel) 2024; 11:476. [PMID: 38790343 PMCID: PMC11117503 DOI: 10.3390/bioengineering11050476] [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: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Organ-on-chip (OOC) technology has gained importance for biomedical studies and drug development. This technology involves microfluidic devices that mimic the structure and function of specific human organs or tissues. OOCs are a promising alternative to traditional cell-based models and animals, as they provide a more representative experimental model of human physiology. By creating a microenvironment that closely resembles in vivo conditions, OOC platforms enable the study of intricate interactions between different cells as well as a better understanding of the underlying mechanisms pertaining to diseases. OOCs can be integrated with other technologies, such as sensors and imaging systems to monitor real-time responses and gather extensive data on tissue behavior. Despite these advances, OOCs for many organs are in their initial stages of development, with several challenges yet to be overcome. These include improving the complexity and maturity of these cellular models, enhancing their reproducibility, standardization, and scaling them up for high-throughput uses. Nonetheless, OOCs hold great promise in advancing biomedical research, drug discovery, and personalized medicine, benefiting human health and well-being. Here, we review several recent OOCs that attempt to overcome some of these challenges. These OOCs with unique applications can be engineered to model organ systems such as the stomach, cornea, blood vessels, and mouth, allowing for analyses and investigations under more realistic conditions. With this, these models can lead to the discovery of potential therapeutic interventions. In this review, we express the significance of the relationship between mucosal tissues and vasculature in organ-on-chip (OOC) systems. This interconnection mirrors the intricate physiological interactions observed in the human body, making it crucial for achieving accurate and meaningful representations of biological processes within OOC models. Vasculature delivers essential nutrients and oxygen to mucosal tissues, ensuring their proper function and survival. This exchange is critical for maintaining the health and integrity of mucosal barriers. This review will discuss the OOCs used to represent the mucosal architecture and vasculature, and it can encourage us to think of ways in which the integration of both can better mimic the complexities of biological systems and gain deeper insights into various physiological and pathological processes. This will help to facilitate the development of more accurate predictive models, which are invaluable for advancing our understanding of disease mechanisms and developing novel therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA; (I.D.); (D.P.R.); (H.A.); (Y.M.)
| |
Collapse
|
23
|
Hall E, Mendiola K, Lightsey NK, Hanjaya-Putra D. Mimicking blood and lymphatic vasculatures using microfluidic systems. BIOMICROFLUIDICS 2024; 18:031502. [PMID: 38726373 PMCID: PMC11081709 DOI: 10.1063/5.0175154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
The role of the circulatory system, containing the blood and lymphatic vasculatures, within the body, has become increasingly focused on by researchers as dysfunction of either of the systems has been linked to serious complications and disease. Currently, in vivo models are unable to provide the sufficient monitoring and level of manipulation needed to characterize the fluidic dynamics of the microcirculation in blood and lymphatic vessels; thus in vitro models have been pursued as an alternative model. Microfluidic devices have the required properties to provide a physiologically relevant circulatory system model for research as well as the experimental tools to conduct more advanced research analyses of microcirculation flow. In this review paper, the physiological behavior of fluid flow and electrical communication within the endothelial cells of the systems are detailed and discussed to highlight their complexities. Cell co-culturing methods and other relevant organ-on-a-chip devices will be evaluated to demonstrate the feasibility and relevance of the in vitro microfluidic model. Microfluidic systems will be determined as a noteworthy model that can display physiologically relevant flow of the cardiovascular and lymphatic systems, which will enable researchers to investigate the systems' prevalence in diseases and identify potential therapeutics.
Collapse
Affiliation(s)
- Eva Hall
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | - N. Keilany Lightsey
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | |
Collapse
|
24
|
Way R, Templeton H, Ball D, Cheng MH, Tobet SA, Chen T. A Microphysiological System for Studying Barrier Health of Live Tissues in Real Time. RESEARCH SQUARE 2024:rs.3.rs-4078220. [PMID: 38659839 PMCID: PMC11042443 DOI: 10.21203/rs.3.rs-4078220/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Epithelial cells create barriers that protect many different components in the body from their external environment. The gut in particular carries bacteria and other infectious agents. A healthy gut epithelial barrier prevents unwanted substances from accessing the underlying lamina propria while maintaining the ability to digest and absorb nutrients. Increased gut barrier permeability, better known as leaky gut, has been linked to several chronic inflammatory diseases. Yet understanding the cause of leaky gut and developing effective interventions are still elusive due to the lack of tools to maintain tissue's physiological environment while elucidating cellular functions under various stimuli ex vivo. This paper presents a microphysiological system capable of recording real-time barrier permeability of mouse gut tissues in a realistic physiological environment over extended durations. Key components of the microphysiological system include a microfluidic chamber designed to hold the live tissue explant and create a sufficient microphysiological environment to maintain tissue viability; proper media composition that preserves a microbiome and creates necessary oxygen gradients across the barrier; integrated sensor electrodes and supporting electronics for acquiring and calculating transepithelial electrical resistance (TEER); and a scalable system architecture to allow multiple chambers running in parallel for increased throughput. The experimental results demonstrate that the system can maintain tissue viability for up to 72 hours. The results also show that the custom-built and integrated TEER sensors are sufficiently sensitive to distinguish differing levels of barrier permeability when treated with collagenase and low pH media compared to control. Permeability variations in tissue explants from different positions in the intestinal tract were also investigated using TEER revealing their disparities in permeability. Finally, the results also quantitatively determine the effect of the muscle layer on total epithelial resistance.
Collapse
Affiliation(s)
- Ryan Way
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Hayley Templeton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Daniel Ball
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Ming-Hao Cheng
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Thomas Chen
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
25
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
26
|
Ponmozhi J, Dhinakaran S, Kocsis D, Iván K, Erdő F. Models for barrier understanding in health and disease in lab-on-a-chips. Tissue Barriers 2024; 12:2221632. [PMID: 37294075 PMCID: PMC11042069 DOI: 10.1080/21688370.2023.2221632] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
The maintenance of body homeostasis relies heavily on physiological barriers. Dysfunction of these barriers can lead to various pathological processes, including increased exposure to toxic materials and microorganisms. Various methods exist to investigate barrier function in vivo and in vitro. To investigate barrier function in a highly reproducible manner, ethically, and high throughput, researchers have turned to non-animal techniques and micro-scale technologies. In this comprehensive review, the authors summarize the current applications of organ-on-a-chip microfluidic devices in the study of physiological barriers. The review covers the blood-brain barrier, ocular barriers, dermal barrier, respiratory barriers, intestinal, hepatobiliary, and renal/bladder barriers under both healthy and pathological conditions. The article then briefly presents placental/vaginal, and tumour/multi-organ barriers in organ-on-a-chip devices. Finally, the review discusses Computational Fluid Dynamics in microfluidic systems that integrate biological barriers. This article provides a concise yet informative overview of the current state-of-the-art in barrier studies using microfluidic devices.
Collapse
Affiliation(s)
- J. Ponmozhi
- Microfluidics Laboratory, Department of Mechanical Engineering, IPS Academy-Institute of Engineering Science, Indore, India
| | - S. Dhinakaran
- The Centre for Fluid Dynamics, Department of Mechanical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Dorottya Kocsis
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Kristóf Iván
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
27
|
Piatnitskaia S, Rafikova G, Bilyalov A, Chugunov S, Akhatov I, Pavlov V, Kzhyshkowska J. Modelling of macrophage responses to biomaterials in vitro: state-of-the-art and the need for the improvement. Front Immunol 2024; 15:1349461. [PMID: 38596667 PMCID: PMC11002093 DOI: 10.3389/fimmu.2024.1349461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/21/2024] [Indexed: 04/11/2024] Open
Abstract
The increasing use of medical implants in various areas of medicine, particularly in orthopedic surgery, oncology, cardiology and dentistry, displayed the limitations in long-term integration of available biomaterials. The effective functioning and successful integration of implants requires not only technical excellence of materials but also consideration of the dynamics of biomaterial interaction with the immune system throughout the entire duration of implant use. The acute as well as long-term decisions about the efficiency of implant integration are done by local resident tissue macrophages and monocyte-derived macrophages that start to be recruited during tissue damage, when implant is installed, and are continuously recruited during the healing phase. Our review summarized the knowledge about the currently used macrophages-based in vitro cells system that include murine and human cells lines and primary ex vivo differentiated macrophages. We provided the information about most frequently examined biomarkers for acute inflammation, chronic inflammation, foreign body response and fibrosis, indicating the benefits and limitations of the model systems. Particular attention is given to the scavenging function of macrophages that controls dynamic composition of peri-implant microenvironment and ensures timely clearance of microorganisms, cytokines, metabolites, extracellular matrix components, dying cells as well as implant debris. We outline the perspective for the application of 3D systems for modelling implant interaction with the immune system in human tissue-specific microenvironment avoiding animal experimentation.
Collapse
Affiliation(s)
- Svetlana Piatnitskaia
- Cell Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Guzel Rafikova
- Additive Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
- Laboratory of Immunology, Institute of Urology and Clinical Oncology, Bashkir State Medical University, Ufa, Russia
| | - Azat Bilyalov
- Additive Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Svyatoslav Chugunov
- Additive Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Iskander Akhatov
- Laboratory of Mathematical modeling, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Valentin Pavlov
- Institute of Urology and Clinical Oncology, Department of Urology, Bashkir State Medical University, Ufa, Russia
| | - Julia Kzhyshkowska
- Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, Mannheim, Germany
| |
Collapse
|
28
|
Shah D, Dave B, Chorawala MR, Prajapati BG, Singh S, M. Elossaily G, Ansari MN, Ali N. An Insight on Microfluidic Organ-on-a-Chip Models for PM 2.5-Induced Pulmonary Complications. ACS OMEGA 2024; 9:13534-13555. [PMID: 38559954 PMCID: PMC10976395 DOI: 10.1021/acsomega.3c10271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024]
Abstract
Pulmonary diseases like asthma, chronic obstructive pulmonary disorder, lung fibrosis, and lung cancer pose a significant burden to global human health. Many of these complications arise as a result of exposure to particulate matter (PM), which has been examined in several preclinical and clinical trials for its effect on several respiratory diseases. Particulate matter of size less than 2.5 μm (PM2.5) has been known to inflict unforeseen repercussions, although data from epidemiological studies to back this are pending. Conventionally utilized two-dimensional (2D) cell culture and preclinical animal models have provided insufficient benefits in emulating the in vivo physiological and pathological pulmonary conditions. Three-dimensional (3D) structural models, including organ-on-a-chip models, have experienced a developmental upsurge in recent times. Lung-on-a-chip models have the potential to simulate the specific features of the lungs. With the advancement of technology, an emerging and advanced technique termed microfluidic organ-on-a-chip has been developed with the aim of identifying the complexity of the respiratory cellular microenvironment of the body. In the present Review, the role of lung-on-a-chip modeling in reproducing pulmonary complications has been explored, with a specific emphasis on PM2.5-induced pulmonary complications.
Collapse
Affiliation(s)
- Disha Shah
- Department
of Pharmacology and Pharmacy Practice, L.
M. College of Pharmacy Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Bhavarth Dave
- Department
of Pharmacology and Pharmacy Practice, L.
M. College of Pharmacy Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Mehul R. Chorawala
- Department
of Pharmacology and Pharmacy Practice, L.
M. College of Pharmacy Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Bhupendra G. Prajapati
- Department
of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education and Research,
Ganpat University, Mehsana, Gujarat 384012, India
| | - Sudarshan Singh
- Office
of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
- Department
of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang
Mai 50200, Thailand
| | - Gehan M. Elossaily
- Department
of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Mohd Nazam Ansari
- Department
of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nemat Ali
- Department
of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
29
|
Amend N, Koller M, Schmitt C, Worek F. Toxicokinetic analysis of the highly toxic nerve agent VX in commercially available multi-organ-chips - Ways to overcome compound absorption. Toxicol Lett 2024; 393:78-83. [PMID: 38311194 DOI: 10.1016/j.toxlet.2024.01.013] [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: 11/28/2023] [Revised: 01/04/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Organ-on-a-chip technology is considered a next-generation platform in pharmacology and toxicology. Nevertheless, this novel technology still faces several challenges concerning the respective materials which are used for these microfluidic devices. Currently available organ-chips are most often based on polydimethylsiloxane (PDMS). However, this material has strong limitations regarding compound binding. The current study investigated options to reduce compound absorption of the highly toxic nerve agent VX (1000 µmol/L) in a commercially available organ-chip. In addition, surface effects on degradation products of VX were investigated. The alternative polymer cyclic olefin copolymers (CoC) showed significantly less compound absorption compared to PDMS. Furthermore, a coating of PDMS- and CoC-based chips was investigated. The biocompatible polymer polyethyleneimine (PEI) successfully modified PDMS and CoC surfaces and further reduced compound absorption. A previously examined VX concentration after 72 h of 141 ± 10 µmol/L VX could be increased to 442 ± 54 µmol/L. Finally, the respective concentrations of VX and degradation products accounted for > 90% of the initial concentration of 1000 µmol/L VX. The currently described surface modification might be a first step towards the optimization of organ-on-a-chip surfaces, facilitating a better comparability of different studies and results.
Collapse
Affiliation(s)
- Niko Amend
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestr. 33, 80336 Munich, Germany.
| | - Marianne Koller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Christian Schmitt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| |
Collapse
|
30
|
Kolanko E, Cargnoni A, Papait A, Silini AR, Czekaj P, Parolini O. The evolution of in vitro models of lung fibrosis: promising prospects for drug discovery. Eur Respir Rev 2024; 33:230127. [PMID: 38232990 DOI: 10.1183/16000617.0127-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/18/2023] [Indexed: 01/19/2024] Open
Abstract
Lung fibrosis is a complex process, with unknown underlying mechanisms, involving various triggers, diseases and stimuli. Different cell types (epithelial cells, endothelial cells, fibroblasts and macrophages) interact dynamically through multiple signalling pathways, including biochemical/molecular and mechanical signals, such as stiffness, affecting cell function and differentiation. Idiopathic pulmonary fibrosis (IPF) is the most common fibrosing interstitial lung disease (fILD), characterised by a notably high mortality. Unfortunately, effective treatments for advanced fILD, and especially IPF and non-IPF progressive fibrosing phenotype ILD, are still lacking. The development of pharmacological therapies faces challenges due to limited knowledge of fibrosis pathogenesis and the absence of pre-clinical models accurately representing the complex features of the disease. To address these challenges, new model systems have been developed to enhance the translatability of preclinical drug testing and bridge the gap to human clinical trials. The use of two- and three-dimensional in vitro cultures derived from healthy or diseased individuals allows for a better understanding of the underlying mechanisms responsible for lung fibrosis. Additionally, microfluidics systems, which replicate the respiratory system's physiology ex vivo, offer promising opportunities for the development of effective therapies, especially for IPF.
Collapse
Affiliation(s)
- Emanuel Kolanko
- Department of Cytophysiology, Katowice Medical University of Silesia in Katowice, Katowice, Poland
- These authors contributed equally
| | - Anna Cargnoni
- Fondazione Poliambulanza Istituto Ospedaliero, Centro di Ricerca E. Menni, Brescia, Italy
- These authors contributed equally
| | - Andrea Papait
- Università Cattolica del Sacro Cuore, Department Life Sciences and Public Health, Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Roma, Italy
| | - Antonietta Rosa Silini
- Fondazione Poliambulanza Istituto Ospedaliero, Centro di Ricerca E. Menni, Brescia, Italy
| | - Piotr Czekaj
- Department of Cytophysiology, Katowice Medical University of Silesia in Katowice, Katowice, Poland
| | - Ornella Parolini
- Università Cattolica del Sacro Cuore, Department Life Sciences and Public Health, Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Roma, Italy
| |
Collapse
|
31
|
dos Santos AEA, Guadalupe JL, Albergaria JDS, Almeida IA, Moreira AMS, Copola AGL, de Araújo IP, de Paula AM, Neves BRA, Santos JPF, da Silva AB, Jorge EC, Andrade LDO. Random cellulose acetate nanofibers: a breakthrough for cultivated meat production. Front Nutr 2024; 10:1297926. [PMID: 38249608 PMCID: PMC10796801 DOI: 10.3389/fnut.2023.1297926] [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: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 01/23/2024] Open
Abstract
Overcoming the challenge of creating thick, tissue-resembling muscle constructs is paramount in the field of cultivated meat production. This study investigates the remarkable potential of random cellulose acetate nanofibers (CAN) as a transformative scaffold for muscle tissue engineering (MTE), specifically in the context of cultivated meat applications. Through a comparative analysis between random and aligned CAN, utilizing C2C12 and H9c2 myoblasts, we unveil the unparalleled capabilities of random CAN in facilitating muscle differentiation, independent of differentiation media, by exploiting the YAP/TAZ-related mechanotransduction pathway. In addition, we have successfully developed a novel process for stacking cell-loaded CAN sheets, enabling the production of a three-dimensional meat product. C2C12 and H9c2 loaded CAN sheets were stacked (up to four layers) to form a ~300-400 μm thick tissue 2 cm in length, organized in a mesh of uniaxial aligned cells. To further demonstrate the effectiveness of this methodology for cultivated meat purposes, we have generated thick and viable constructs using chicken muscle satellite cells (cSCs) and random CAN. This groundbreaking discovery offers a cost-effective and biomimetic solution for cultivating and differentiating muscle cells, forging a crucial link between tissue engineering and the pursuit of sustainable and affordable cultivated meat production.
Collapse
Affiliation(s)
- Ana Elisa Antunes dos Santos
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jorge Luís Guadalupe
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliano Douglas Silva Albergaria
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Itallo Augusto Almeida
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Amanda Maria Siqueira Moreira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Aline Gonçalves Lio Copola
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Isabella Paula de Araújo
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Ana Maria de Paula
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bernardo Ruegger Almeida Neves
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João Paulo Ferreira Santos
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Aline Bruna da Silva
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luciana de Oliveira Andrade
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
32
|
Rama Varma A, Fathi P. Vascularized microfluidic models of major organ structures and cancerous tissues. BIOMICROFLUIDICS 2023; 17:061502. [PMID: 38074952 PMCID: PMC10703512 DOI: 10.1063/5.0159800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/13/2023] [Indexed: 10/16/2024]
Abstract
Organ-on-a-chip devices are powerful modeling systems that allow researchers to recapitulate the in vivo structures of organs as well as the physiological conditions those tissues are subject to. These devices are useful tools in modeling not only the behavior of a healthy organ but also in modeling disease pathology or the effects of specific drugs. The incorporation of fluidic flow is of great significance in these devices due to the important roles of physiological fluid flows in vivo. Recent developments in the field have led to the production of vascularized organ-on-a-chip devices, which can more accurately reproduce the conditions observed in vivo by recapitulating the vasculature of the organ concerned. This review paper will provide a brief overview of the history of organ-on-a-chip devices, before discussing developments in the production of vascularized organs-on-chips, and the implications these developments hold for the future of the field.
Collapse
Affiliation(s)
- Anagha Rama Varma
- Unit for NanoEngineering and MicroPhysiological Systems, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Parinaz Fathi
- Unit for NanoEngineering and MicroPhysiological Systems, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
33
|
Kim H, Kim GS, Hyun SH, Kim E. Advancements in 2D and 3D In Vitro Models for Studying Neuromuscular Diseases. Int J Mol Sci 2023; 24:17006. [PMID: 38069329 PMCID: PMC10707046 DOI: 10.3390/ijms242317006] [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: 10/28/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Neuromuscular diseases (NMDs) are a genetically or clinically heterogeneous group of diseases that involve injury or dysfunction of neuromuscular tissue components, including peripheral motor neurons, skeletal muscles, and neuromuscular junctions. To study NMDs and develop potential therapies, remarkable progress has been made in generating in vitro neuromuscular models using engineering approaches to recapitulate the complex physical and biochemical microenvironments of 3D human neuromuscular tissues. In this review, we discuss recent studies focusing on the development of in vitro co-culture models of human motor neurons and skeletal muscles, with the pros and cons of each approach. Furthermore, we explain how neuromuscular in vitro models recapitulate certain aspects of specific NMDs, including amyotrophic lateral sclerosis and muscular dystrophy. Research on neuromuscular organoids (NMO) will continue to co-develop to better mimic tissues in vivo and will provide a better understanding of the development of the neuromuscular tissue, mechanisms of NMD action, and tools applicable to preclinical studies, including drug screening and toxicity tests.
Collapse
Affiliation(s)
- Haneul Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea;
- Institute for Stem Cell & Regenerative Medicine, Chungbuk National University, Chengju 28644, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Eunhye Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| |
Collapse
|
34
|
Mironiuk-Puchalska E, Karatsai O, Żuchowska A, Wróblewski W, Borys F, Lehka L, Rędowicz MJ, Koszytkowska-Stawińska M. Development of 5-fluorouracil-dichloroacetate mutual prodrugs as anticancer agents. Bioorg Chem 2023; 140:106784. [PMID: 37639758 DOI: 10.1016/j.bioorg.2023.106784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
5-Fluorouracil (5-FU) is one of the most widely applied chemotherapeutic agents with a broad spectrum of activity. However, despite this versatile activity, its use poses many limitations. Herein, novel derivatives of 5-FU and dichloroacetic acid have been designed and synthesized as a new type of codrugs, also known as mutual prodrugs, to overcome the drawbacks of 5-FU and enhance its therapeutic efficiency. The stability of the obtained compounds has been tested at various pH values using different analytical techniques, namely HPLC and potentiometry. The antiproliferative activity of the new 5-FU derivatives was assessed in vitro on SK-MEL-28 and WM793 human melanoma cell lines in 2D culture as well as on A549 human lung carcinoma, MDA-MB-231 breast adenocarcinoma, LL24 normal lung tissue, and HMF normal breast tissue as a multicellular 3D spheroid model cultured in standard (static) conditions and with the use of microfluidic systems, which to a great extent resembles the in vivo environment. In all cases, new mutual prodrugs showed a higher cytotoxic activity toward cancer models and lower to normal cell models than the parent 5-FU itself.
Collapse
Affiliation(s)
- Ewa Mironiuk-Puchalska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland.
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Agnieszka Żuchowska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Wojciech Wróblewski
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Filip Borys
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | | |
Collapse
|
35
|
Amend N, Koller M, Schmitt C, Worek F, Wille T. The suitability of a polydimethylsiloxane-based (PDMS) microfluidic two compartment system for the toxicokinetic analysis of organophosphorus compounds. Toxicol Lett 2023; 388:24-29. [PMID: 37827339 DOI: 10.1016/j.toxlet.2023.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023]
Abstract
Organ-on-a-chip platforms are an emerging technology in experimental and regulatory toxicology (species-specific differences, ethical considerations). They address gaps between in vivo and in vitro models. However, there are still certain limitations considering material, setup and applicability. The current study examined the suitability of a commercially available polydimethylsiloxane-based (PDMS) organ-chip for the toxicokinetic characterization of the highly toxic nerve agent VX and the organophosphate pesticide parathion. The respective concentrations of 1000 µmol/L and 100 µmol/L VX and parathion were chosen deliberately high in order to study concentrations even if high compound absorption by PDMS might occur. Neuronal and liver spheroids, totaling 2 × 106 cells were used to study concentration changes of VX and parathion. In addition, VX enantiomers were quantified. The current study suggests a significant absorption of VX, respectively parathion by PDMS. This might require future investigation of alternative materials or coatings to limit absorption for organophosphorus compounds in toxicokinetic studies.
Collapse
Affiliation(s)
- Niko Amend
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany.
| | - Marianne Koller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Christian Schmitt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany; Department F, Medical CBRN Defence, Bundeswehr Medical Academy, Ingolstädter Str 240, Munich 80939, Germany
| |
Collapse
|
36
|
Thakar RG, Fenton KN. Bioethical implications of organ-on-a-chip on modernizing drug development. Artif Organs 2023; 47:1553-1558. [PMID: 37578206 PMCID: PMC10615722 DOI: 10.1111/aor.14620] [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] [Indexed: 08/15/2023]
Abstract
Organ-on-chips are three-dimensional microdevices that emulate the structure, functionality, and behavior of specific tissues or organs using human cells. Combining organoids with microfabricated fluidic channels and microelectronics, these systems offer a promising platform for studying disease mechanisms, drug responses, and tissue performance. By replicating the in vivo microenvironment, these devices can recreate complex cell interactions in controlled conditions and facilitate research in various fields, including drug toxicity and efficacy studies, biochemical analysis, and disease pathogenesis. Integrating human induced pluripotent stem cells further enhances their applicability, thereby enabling patient-specific disease modeling for precision medicine. Although challenges like economy-of-scale, multichip integration, and regulatory compliance exist, advances in this modular technology show promise for lowering drug development costs, improving reproducibility, and reducing the reliance on animal testing. The ethical landscape surrounding organ-on-chip usage presents both benefits and concerns. While these chips offer an alternative to animal testing and potential cost savings, they raise ethical considerations related to community engagement, informed consent, and the need for standardized guidelines. Ensuring public acceptance and involvement in decision-making is vital to address misinformation and mistrust. Furthermore, personalized medicine models using patient-derived cells demand careful consideration of potential ethical dilemmas, such as modeling physiological functions of fetuses or brains and determining the extent of protection for these models. To achieve the full potential of organ-on-a-chip models, collaboration between scientists, ethicists, and regulators is essential to fulfil the promise of transforming drug development, advancing personalized medicine, and contributing to a more ethical and efficient biomedical research landscape.
Collapse
Affiliation(s)
- Rahul G. Thakar
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kathleen N. Fenton
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Department of Bioethics, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
37
|
Hu C, Yang W. Alternatives to animal models to study bacterial infections. Folia Microbiol (Praha) 2023; 68:703-739. [PMID: 37632640 DOI: 10.1007/s12223-023-01084-6] [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: 04/14/2023] [Accepted: 08/02/2023] [Indexed: 08/28/2023]
Abstract
Animal testing has made a significant and unequalled contribution to important discoveries and advancements in the fields of research, medicine, vaccine development, and drug discovery. Each year, millions of animals are sacrificed for various experiments, and this is an ongoing process. However, the debate on the ethical and sensible usage of animals in in vivo experimentation is equally important. The need to explore and adopt newer alternatives to animals so as to comply with the goal of reduce, refine, and replace needs attention. Besides the ever-increasing debate on ethical issues, animal research has additional drawbacks (need of trained labour, requirement of breeding area, lengthy protocols, high expenses, transport barriers, difficulty to extrapolate data from animals to humans, etc.). With this scenario, the present review has been framed to give a comprehensive insight into the possible alternative options worth exploring in this direction especially targeting replacements for animal models of bacterial infections. There have been some excellent reviews discussing on the alternate methods for replacing and reducing animals in drug research. However, reviews that discuss the replacements in the field of medical bacteriology with emphasis on animal bacterial infection models are purely limited. The present review discusses on the use of (a) non-mammalian models and (b) alternative systems such as microfluidic chip-based models and microdosing aiming to give a detailed insight into the prospects of these alternative platforms to reduce the number of animals being used in infection studies. This would enlighten the scientific community working in this direction to be well acquainted with the available new approaches and alternatives so that the 3R strategy can be successfully implemented in the field of antibacterial drug research and testing.
Collapse
Affiliation(s)
- Chengming Hu
- Queen Mary College, Nanchang University, Nanchang, China
| | - Wenlong Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
38
|
van Rijt A, Stefanek E, Valente K. Preclinical Testing Techniques: Paving the Way for New Oncology Screening Approaches. Cancers (Basel) 2023; 15:4466. [PMID: 37760435 PMCID: PMC10526899 DOI: 10.3390/cancers15184466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Prior to clinical trials, preclinical testing of oncology drug candidates is performed by evaluating drug candidates with in vitro and in vivo platforms. For in vivo testing, animal models are used to evaluate the toxicity and efficacy of drug candidates. However, animal models often display poor translational results as many drugs that pass preclinical testing fail when tested with humans, with oncology drugs exhibiting especially poor acceptance rates. The FDA Modernization Act 2.0 promotes alternative preclinical testing techniques, presenting the opportunity to use higher complexity in vitro models as an alternative to in vivo testing, including three-dimensional (3D) cell culture models. Three-dimensional tissue cultures address many of the shortcomings of 2D cultures by more closely replicating the tumour microenvironment through a combination of physiologically relevant drug diffusion, paracrine signalling, cellular phenotype, and vascularization that can better mimic native human tissue. This review will discuss the common forms of 3D cell culture, including cell spheroids, organoids, organs-on-a-chip, and 3D bioprinted tissues. Their advantages and limitations will be presented, aiming to discuss the use of these 3D models to accurately represent human tissue and as an alternative to animal testing. The use of 3D culture platforms for preclinical drug development is expected to accelerate as these platforms continue to improve in complexity, reliability, and translational predictivity.
Collapse
Affiliation(s)
- Antonia van Rijt
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Evan Stefanek
- VoxCell BioInnovation Inc., Victoria, BC V8T 5L2, Canada;
| | - Karolina Valente
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada;
| |
Collapse
|
39
|
Gil JF, Moura CS, Silverio V, Gonçalves G, Santos HA. Cancer Models on Chip: Paving the Way to Large-Scale Trial Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300692. [PMID: 37103886 DOI: 10.1002/adma.202300692] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/05/2023] [Indexed: 06/19/2023]
Abstract
Cancer kills millions of individuals every year all over the world (Global Cancer Observatory). The physiological and biomechanical processes underlying the tumor are still poorly understood, hindering researchers from creating new, effective therapies. Inconsistent results of preclinical research, in vivo testing, and clinical trials decrease drug approval rates. 3D tumor-on-a-chip (ToC) models integrate biomaterials, tissue engineering, fabrication of microarchitectures, and sensory and actuation systems in a single device, enabling reliable studies in fundamental oncology and pharmacology. This review includes a critical discussion about their ability to reproduce the tumor microenvironment (TME), the advantages and drawbacks of existing tumor models and architectures, major components and fabrication techniques. The focus is on current materials and micro/nanofabrication techniques used to manufacture reliable and reproducible microfluidic ToC models for large-scale trial applications.
Collapse
Affiliation(s)
- João Ferreira Gil
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, Marinha Grande, 2430-028, Portugal
- INESC Microsistemas e Nanotecnologias (INESC MN), Rua Alves Redol 9, Lisbon, 1000-029, Portugal
- TEMA, Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Carla Sofia Moura
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, Marinha Grande, 2430-028, Portugal
- Polytechnic Institute of Coimbra, Applied Research Institute, Coimbra, 3045-093, Portugal
| | - Vania Silverio
- INESC Microsistemas e Nanotecnologias (INESC MN), Rua Alves Redol 9, Lisbon, 1000-029, Portugal
- Department of Physics, Instituto Superior Técnico, Lisbon, 1049-001, Portugal
- Associate Laboratory Institute for Health and Bioeconomy - i4HB, Lisbon, Portugal
| | - Gil Gonçalves
- TEMA, Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
- Intelligent Systems Associate Laboratory (LASI), Aveiro, 3810-193, Portugal
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- W.J. Korf Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| |
Collapse
|
40
|
Padmyastuti A, Sarmiento MG, Dib M, Ehrhardt J, Schoon J, Somova M, Burchardt M, Roennau C, Pinto PC. Microfluidic-based prostate cancer model for investigating the secretion of prostate-specific antigen and microRNAs in vitro. Sci Rep 2023; 13:11623. [PMID: 37468746 DOI: 10.1038/s41598-023-38834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023] Open
Abstract
The study of prostate cancer in vitro relies on established cell lines that lack important physiological characteristics, such as proper polarization and expression of relevant biomarkers. Microphysiological systems (MPS) can replicate cancer microenvironments and lead to cellular phenotypic changes that better represent organ physiology in vitro. In this study, we developed an MPS model comprising conventional prostate cancer cells to evaluate their activity under dynamic culture conditions. Androgen-sensitive (LNCaP) and androgen-insensitive (PC3) cells were grown in conventional and 3D cultures, both static and dynamic. Cell morphology, the secretion of prostate-specific antigen, and the expression of key prostate markers and microRNAs were analyzed. LNCaP formed spheroids in 3D and MPS cultures, with morphological changes supported by the upregulation of cytokeratins and adhesion proteins. LNCaP also maintained a constant prostate-specific antigen secretion in MPS. PC3 cells did not develop complex structures in 3D and MPS cultures. PSA expression at the gene level was downregulated in LNCaP-MPS and considerably upregulated in PC3-MPS. MicroRNA expression was altered by the 3D static and dynamic culture, both intra- and extracellularly. MicroRNAs associated with prostate cancer progression were mostly upregulated in LNCaP-MPS. Overall dynamic cell culture substantially altered the morphology and expression of LNCaP cells, arguably augmenting their prostate cancer phenotype. This novel approach demonstrates that microRNA expression in prostate cancer cells is sensitive to external stimuli and that MPS can effectively promote important physiological changes in conventional prostate cancer models.
Collapse
Affiliation(s)
- Adventina Padmyastuti
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Marina Garcia Sarmiento
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Maria Dib
- Department of Ear, Nose and Throat Surgery, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Jens Ehrhardt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Janosch Schoon
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Fleichmannstraße 8, 17475, Greifswald, Germany
| | - Maryna Somova
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Cindy Roennau
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Pedro Caetano Pinto
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany.
| |
Collapse
|
41
|
Sharma S, Khan Q, Schreurs OJF, Sapkota D, Samuelsen JT. Investigation of biological effects of HEMA in 3D-organotypic co-culture models of normal and malignant oral keratinocytes. Biomater Investig Dent 2023; 10:2234400. [PMID: 37456807 PMCID: PMC10348043 DOI: 10.1080/26415275.2023.2234400] [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: 02/14/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Several in vitro studies utilizing 2-dimensional (2D) cell culture systems have linked 2-hydroxyethyl methacrylate (HEMA) with cytotoxic effects in oral mucosa and dental pulp cells. Although such studies are invaluable in dissecting the cellular and molecular effects of HEMA, there is a growing interest in the utilization of appropriate 3-dimensional (3D) models that mimic the structure of oral mucosa. Using a previously characterized 3D-organotypic co-culture model, this study aimed to investigate the cellular and molecular effects of HEMA on a 3D-co-culture model consisting of primary normal oral keratinocyte (NOK) grown directly on top of collagen I gel containing primary oral fibroblasts (NOF). The second aim was to examine the suitability of a 3D-co-culture system consisting of oral squamous cell carcinoma (OSCC) cells as a model system to investigate the biological effects of HEMA. We demonstrated that HEMA treatment led to reduced viability of NOK, NOF and OSCC-cell lines in 2D-culture. The keratinocytes in 3D-co-cultures of NOK and OSCC-cells reacted similarly with respect to cell proliferation and activation of autophagy flux, to HEMA treatment. Nevertheless, NOK was found to be more susceptible to apoptosis following HEMA treatment than OSCC in 3D-co-cultures. These results indicate that 3D-organotypic co-cultures of NOK might represent an appropriate model system for the investigation of the biological effects of HEMA and other dental biomaterials. Given the challenges in obtaining primary cultures of NOK and issues associated with their rapid differentiation in culture, the possible use of OSCC cells as an alternative to NOK for 3D models represents an area for future research.
Collapse
Affiliation(s)
- Sunita Sharma
- Nordic Institute of Dental Materials, Oslo, Norway
- Christiania Dental Clinic, Malo Dental, Oslo, Norway
| | - Qalbi Khan
- Department of Oral Biology, University of Oslo, Oslo, Blindern, Norway
| | | | - Dipak Sapkota
- Department of Oral Biology, University of Oslo, Oslo, Blindern, Norway
| | | |
Collapse
|
42
|
Sahoo DK, Martinez MN, Dao K, Gabriel V, Zdyrski C, Jergens AE, Atherly T, Iennarella-Servantez CA, Burns LE, Schrunk D, Volpe DA, Allenspach K, Mochel JP. Canine Intestinal Organoids as a Novel In Vitro Model of Intestinal Drug Permeability: A Proof-of-Concept Study. Cells 2023; 12:cells12091269. [PMID: 37174669 PMCID: PMC10177590 DOI: 10.3390/cells12091269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
A key component of efforts to identify the biological and drug-specific aspects contributing to therapeutic failure or unexpected exposure-associated toxicity is the study of drug-intestinal barrier interactions. While methods supporting such assessments are widely described for human therapeutics, relatively little information is available for similar evaluations in support of veterinary pharmaceuticals. There is, therefore, a critical need to develop novel approaches for evaluating drug-gut interactions in veterinary medicine. Three-dimensional (3D) organoids can address these difficulties in a reasonably affordable system that circumvents the need for more invasive in vivo assays in live animals. However, a first step in developing such systems is understanding organoid interactions in a 2D monolayer. Given the importance of orally administered medications for meeting the therapeutic need of companion animals, we demonstrate growth conditions under which canine-colonoid-derived intestinal epithelial cells survive, mature, and differentiate into confluent cell systems with high monolayer integrity. We further examine the applicability of this canine-colonoid-derived 2D model to assess the permeability of three structurally diverse, passively absorbed β-blockers (e.g., propranolol, metoprolol, and atenolol). Both the absorptive and secretive apparent permeability (Papp) of these drugs at two different pH conditions were evaluated in canine-colonoid-derived monolayers and compared with that of Caco-2 cells. This proof-of-concept study provides promising preliminary results with regard to the utility of canine-derived organoid monolayers for species-specific assessments of therapeutic drug passive permeability.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Marilyn N Martinez
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, Food and Drug Administration, Rockville, MD 20852, USA
| | - Kimberly Dao
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
| | - Vojtech Gabriel
- Department of Biomedical Sciences, SMART Pharmacology, Iowa State University, Ames, IA 50011, USA
| | - Christopher Zdyrski
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
- Department of Biomedical Sciences, SMART Pharmacology, Iowa State University, Ames, IA 50011, USA
| | - Albert E Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Todd Atherly
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
| | | | - Laura E Burns
- Veterinary Diagnostic Laboratory, Iowa State University, Ames, IA 50011, USA
| | - Dwayne Schrunk
- Veterinary Diagnostic Laboratory, Iowa State University, Ames, IA 50011, USA
| | - Donna A Volpe
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20852, USA
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
| | - Jonathan P Mochel
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
- Department of Biomedical Sciences, SMART Pharmacology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
43
|
Xue B, Schüler J, Harrod CM, Lashuk K, Bomya Z, Hribar KC. A Novel Hydrogel-Based 3D In Vitro Tumor Panel of 30 PDX Models Incorporates Tumor, Stromal and Immune Cell Compartments of the TME for the Screening of Oncology and Immuno-Therapies. Cells 2023; 12:1145. [PMID: 37190054 PMCID: PMC10137152 DOI: 10.3390/cells12081145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Human-relevant systems that mimic the 3D tumor microenvironment (TME), particularly the complex mechanisms of immuno-modulation in the tumor stroma, in a reproducible and scalable format are of high interest for the drug discovery industry. Here, we describe a novel 3D in vitro tumor panel comprising 30 distinct PDX models covering a range of histotypes and molecular subtypes and cocultured with fibroblasts and PBMCs in planar (flat) extracellular matrix hydrogels to reflect the three compartments of the TME-tumor, stroma, and immune cells. The panel was constructed in a 96-well plate format and assayed tumor size, tumor killing, and T-cell infiltration using high-content image analysis after 4 days of treatment. We screened the panel first against the chemotherapy drug Cisplatin to demonstrate feasibility and robustness, and subsequently assayed immuno-oncology agents Solitomab (CD3/EpCAM bispecific T-cell engager) and the immune checkpoint inhibitors (ICIs) Atezolizumab (anti-PDL1), Nivolumab (anti-PD1) and Ipilimumab (anti-CTLA4). Solitomab displayed a strong response across many PDX models in terms of tumor reduction and killing, allowing for its subsequent use as a positive control for ICIs. Interestingly, Atezolizumab and Nivolumab demonstrated a mild response compared to Ipilimumab in a subset of models from the panel. We later determined that PBMC spatial proximity in the assay setup was important for the PD1 inhibitor, hypothesizing that both duration and concentration of antigen exposure may be critical. The described 30-model panel represents a significant advancement toward screening in vitro models of the tumor microenvironment that include tumor, fibroblast, and immune cell populations in an extracellular matrix hydrogel, with robust and standardized high content image analysis in a planar hydrogel. The platform is aimed at rapidly screening various combinations and novel agents and forming a critical conduit to the clinic, thus accelerating drug discovery for the next generation of therapeutics.
Collapse
Affiliation(s)
- Bin Xue
- Cypre, Inc., South San Francisco, CA 94080, USA
| | - Julia Schüler
- Charles River Discovery Research Services Germany GmbH, 79108 Freiburg, Germany
| | | | - Kanstantsin Lashuk
- Charles River Discovery Research Services Germany GmbH, 79108 Freiburg, Germany
| | - Zoji Bomya
- Cypre, Inc., South San Francisco, CA 94080, USA
| | | |
Collapse
|
44
|
Manduca N, Maccafeo E, De Maria R, Sistigu A, Musella M. 3D cancer models: One step closer to in vitro human studies. Front Immunol 2023; 14:1175503. [PMID: 37114038 PMCID: PMC10126361 DOI: 10.3389/fimmu.2023.1175503] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer immunotherapy is the great breakthrough in cancer treatment as it displayed prolonged progression-free survival over conventional therapies, yet, to date, in only a minority of patients. In order to broad cancer immunotherapy clinical applicability some roadblocks need to be overcome, first among all the lack of preclinical models that faithfully depict the local tumor microenvironment (TME), which is known to dramatically affect disease onset, progression and response to therapy. In this review, we provide the reader with a detailed overview of current 3D models developed to mimick the complexity and the dynamics of the TME, with a focus on understanding why the TME is a major target in anticancer therapy. We highlight the advantages and translational potentials of tumor spheroids, organoids and immune Tumor-on-a-Chip models in disease modeling and therapeutic response, while outlining pending challenges and limitations. Thinking forward, we focus on the possibility to integrate the know-hows of micro-engineers, cancer immunologists, pharmaceutical researchers and bioinformaticians to meet the needs of cancer researchers and clinicians interested in using these platforms with high fidelity for patient-tailored disease modeling and drug discovery.
Collapse
Affiliation(s)
- Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ‘A. Gemelli’ - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
45
|
Valiei A, Aminian-Dehkordi J, Mofrad MRK. Gut-on-a-chip models for dissecting the gut microbiology and physiology. APL Bioeng 2023; 7:011502. [PMID: 36875738 PMCID: PMC9977465 DOI: 10.1063/5.0126541] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/23/2023] [Indexed: 03/04/2023] Open
Abstract
Microfluidic technologies have been extensively investigated in recent years for developing organ-on-a-chip-devices as robust in vitro models aiming to recapitulate organ 3D topography and its physicochemical cues. Among these attempts, an important research front has focused on simulating the physiology of the gut, an organ with a distinct cellular composition featuring a plethora of microbial and human cells that mutually mediate critical body functions. This research has led to innovative approaches to model fluid flow, mechanical forces, and oxygen gradients, which are all important developmental cues of the gut physiological system. A myriad of studies has demonstrated that gut-on-a-chip models reinforce a prolonged coculture of microbiota and human cells with genotypic and phenotypic responses that closely mimic the in vivo data. Accordingly, the excellent organ mimicry offered by gut-on-a-chips has fueled numerous investigations on the clinical and industrial applications of these devices in recent years. In this review, we outline various gut-on-a-chip designs, particularly focusing on different configurations used to coculture the microbiome and various human intestinal cells. We then elaborate on different approaches that have been adopted to model key physiochemical stimuli and explore how these models have been beneficial to understanding gut pathophysiology and testing therapeutic interventions.
Collapse
Affiliation(s)
- Amin Valiei
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California 94720, USA
| | - Javad Aminian-Dehkordi
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California 94720, USA
| | | |
Collapse
|
46
|
Soto Veliz D, Lin K, Sahlgren C. Organ-on-a-chip technologies for biomedical research and drug development: A focus on the vasculature. SMART MEDICINE 2023; 2:e20220030. [PMID: 37089706 PMCID: PMC7614466 DOI: 10.1002/smmd.20220030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/20/2023] [Indexed: 04/25/2023]
Abstract
Current biomedical models fail to replicate the complexity of human biology. Consequently, almost 90% of drug candidates fail during clinical trials after decades of research and billions of investments in drug development. Despite their physiological similarities, animal models often misrepresent human responses, and instead, trigger ethical and societal debates regarding their use. The overall aim across regulatory entities worldwide is to replace, reduce, and refine the use of animal experimentation, a concept known as the Three Rs principle. In response, researchers develop experimental alternatives to improve the biological relevance of in vitro models through interdisciplinary approaches. This article highlights the emerging organ-on-a-chip technologies, also known as microphysiological systems, with a focus on models of the vasculature. The cardiovascular system transports all necessary substances, including drugs, throughout the body while in charge of thermal regulation and communication between other organ systems. In addition, we discuss the benefits, limitations, and challenges in the widespread use of new biomedical models. Coupled with patient-derived induced pluripotent stem cells, organ-on-a-chip technologies are the future of drug discovery, development, and personalized medicine.
Collapse
Affiliation(s)
- Diosangeles Soto Veliz
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Kai‐Lan Lin
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Cecilia Sahlgren
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoventhe Netherlands
| |
Collapse
|
47
|
Sønstevold L, Czerkies M, Escobedo-Cousin E, Blonski S, Vereshchagina E. Application of Polymethylpentene, an Oxygen Permeable Thermoplastic, for Long-Term on-a-Chip Cell Culture and Organ-on-a-Chip Devices. MICROMACHINES 2023; 14:532. [PMID: 36984939 PMCID: PMC10053898 DOI: 10.3390/mi14030532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
The applicability of a gas-permeable, thermoplastic material polymethylpentene (PMP) was investigated, experimentally and analytically, for organ-on-a-chip (OoC) and long-term on-a-chip cell cultivation applications. Using a sealed culture chamber device fitted with oxygen sensors, we tested and compared PMP to commonly used glass and polydimethylsiloxane (PDMS). We show that PMP and PDMS have comparable performance for oxygen supply during 4 days culture of epithelial (A549) cells with oxygen concentration stabilizing at 16%, compared with glass control where it decreases to 3%. For the first time, transmission light images of cells growing on PMP were obtained, demonstrating that the optical properties of PMP are suitable for non-fluorescent, live cell imaging. Following the combined transmission light imaging and calcein-AM staining, cell adherence, proliferation, morphology, and viability of A549 cells were shown to be similar on PMP and glass coated with poly-L-lysine. In contrast to PDMS, we demonstrate that a film of PMP as thin as 0.125 mm is compatible with high-resolution confocal microscopy due to its excellent optical properties and mechanical stiffness. PMP was also found to be fully compatible with device sterilization, cell fixation, cell permeabilization and fluorescent staining. We envision this material to extend the range of possible microfluidic applications beyond the current state-of-the-art, due to its beneficial physical properties and suitability for prototyping by different methods. The integrated device and measurement methodology demonstrated in this work are transferrable to other cell-based studies and life-sciences applications.
Collapse
Affiliation(s)
- Linda Sønstevold
- SINTEF Digital, Department of Smart Sensors and Microsystems, Gaustadalléen 23C, 0373 Oslo, Norway
| | - Maciej Czerkies
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego St. 5B, 02-106 Warsaw, Poland
| | - Enrique Escobedo-Cousin
- SINTEF Digital, Department of Smart Sensors and Microsystems, Gaustadalléen 23C, 0373 Oslo, Norway
| | - Slawomir Blonski
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego St. 5B, 02-106 Warsaw, Poland
| | - Elizaveta Vereshchagina
- SINTEF Digital, Department of Smart Sensors and Microsystems, Gaustadalléen 23C, 0373 Oslo, Norway
| |
Collapse
|
48
|
Dogan AA, Dufva M. Heterogenous morphogenesis of Caco-2 cells reveals that flow induces three-dimensional growth and maturation at high initial seeding cell densities. Biotechnol Bioeng 2023; 120:1667-1677. [PMID: 36815727 DOI: 10.1002/bit.28362] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/25/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Here, we introduce a customized hanging insert fitting a six-well plate to culture Caco-2 cells on hydrogel membranes under flow conditions. The cells are cultured in the apical channel-like chamber, which provides about 1.3 dyn/cm2 shear, while the basolateral chamber is mixed when the device is rocked. The device was tested by investigating the functional impact of the initial seeding density in combination with flow applied at confluency. The low seeding density cultures grew in two dimensional (2D) irrespective of the flow. Flow and higher seeding density resulted in a mixture of three dimensional (3D) structures and 2D layers. Static culture and high cell seeding density resulted in 2D layers. The flow increased the height and ZO-1 expression of cells in 2D layers, which correlated with an improved barrier function. Cultures with 3D structures had higher ZO-1 expression than 2D cultures, but this did not correlate with an increased barrier function. 2D monolayers in static and dynamic cultures had similar morphology and heterogeneity in the expression of Mucin-2 and Villin, while the 3D structures had generally higher expression of these markers. The result shows that the cell density and flow determine 3D growth and that the highest barrier function was obtained with low-density cultures and flow.
Collapse
Affiliation(s)
- Asli Aybike Dogan
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Martin Dufva
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
49
|
Chen Z, Huang J, Zhang J, Xu Z, Li Q, Ouyang J, Yan Y, Sun S, Ye H, Wang F, Zhu J, Wang Z, Chao J, Pu Y, Gu Z. A storm in a teacup -- A biomimetic lung microphysiological system in conjunction with a deep-learning algorithm to monitor lung pathological and inflammatory reactions. Biosens Bioelectron 2023; 219:114772. [PMID: 36272347 DOI: 10.1016/j.bios.2022.114772] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/07/2022] [Accepted: 09/28/2022] [Indexed: 10/06/2022]
Abstract
Creating a biomimetic in vitro lung model to recapitulate the infection and inflammatory reactions has been an important but challenging task for biomedical researchers. The 2D based cell culture models - culturing of lung epithelium - have long existed but lack multiple key physiological conditions, such as the involvement of different types of immune cells and the creation of connected lung models to study viral or bacterial infection between different individuals. Pioneers in organ-on-a-chip research have developed lung alveoli-on-a-chip and connected two lung chips with direct tubing and flow. Although this model provides a powerful tool for lung alveolar disease modeling, it still lacks interactions among immune cells, such as macrophages and monocytes, and the mimic of air flow and aerosol transmission between lung-chips is missing. Here, we report the development of an improved human lung physiological system (Lung-MPS) with both alveolar and pulmonary bronchial chambers that permits the integration of multiple immune cells into the system. We observed amplified inflammatory signals through the dynamic interactions among macrophages, epithelium, endothelium, and circulating monocytes. Furthermore, an integrated microdroplet/aerosol transmission system was fabricated and employed to study the propagation of pseudovirus particles containing microdroplets in integrated Lung-MPSs. Finally, a deep-learning algorithm was developed to characterize the activation of cells in this Lung-MPS. This Lung-MPS could provide an improved and more biomimetic sensory system for the study of COVID-19 and other high-risk infectious lung diseases.
Collapse
Affiliation(s)
- Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou#2, Nanjing, Jiangsu, 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Jie Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210000, China; Department of Respiratory and Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jing Zhang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Zikang Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou#2, Nanjing, Jiangsu, 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou#2, Nanjing, Jiangsu, 210096, China
| | - Jun Ouyang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Yuchuan Yan
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Shiqi Sun
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Huan Ye
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Fei Wang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Jianfeng Zhu
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Zhangyan Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210000, China
| | - Jie Chao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210000, China.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210000, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou#2, Nanjing, Jiangsu, 210096, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210000, China.
| |
Collapse
|
50
|
Sano T, Nakajima T, Senda KA, Nakano S, Yamato M, Ikeda Y, Zeng H, Kawabe JI, Matsunaga YT. Image-based crosstalk analysis of cell-cell interactions during sprouting angiogenesis using blood-vessel-on-a-chip. Stem Cell Res Ther 2022; 13:532. [PMID: 36575469 PMCID: PMC9795717 DOI: 10.1186/s13287-022-03223-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Sprouting angiogenesis is an important mechanism for morphogenetic phenomena, including organ development, wound healing, and tissue regeneration. In regenerative medicine, therapeutic angiogenesis is a clinical solution for recovery from ischemic diseases. Mesenchymal stem cells (MSCs) have been clinically used given their pro-angiogenic effects. MSCs are reported to promote angiogenesis by differentiating into pericytes or other vascular cells or through cell-cell communication using multiple protein-protein interactions. However, how MSCs physically contact and move around ECs to keep the sprouting angiogenesis active remains unknown. METHODS We proposed a novel framework of EC-MSC crosstalk analysis using human umbilical vein endothelial cells (HUVECs) and MSCs obtained from mice subcutaneous adipose tissue on a 3D in vitro model, microvessel-on-a-chip, which allows cell-to-tissue level study. The microvessels were fabricated and cultured for 10 days in a collagen matrix where MSCs were embedded. RESULTS Immunofluorescence imaging using a confocal laser microscope showed that MSCs smoothed the surface of the microvessel and elongated the angiogenic sprouts by binding to the microvessel's specific microstructures. Additionally, three-dimensional modeling of HUVEC-MSC intersections revealed that MSCs were selectively located around protrusions or roots of angiogenic sprouts, whose surface curvature was excessively low or high, respectively. CONCLUSIONS The combination of our microvessel-on-a-chip system for 3D co-culture and image-based crosstalk analysis demonstrated that MSCs are selectively localized to concave-convex surfaces on scaffold structures and that they are responsible for the activation and stabilization of capillary vessels.
Collapse
Affiliation(s)
- Takanori Sano
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Tadaaki Nakajima
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan ,grid.268441.d0000 0001 1033 6139Department of Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027 Japan
| | - Koharu Alicia Senda
- Hiroo Gakuen Junior and Senior High School, 5-1-14 Minami Azabu, Minato-ku, Tokyo, 106-0047 Japan
| | - Shizuka Nakano
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Mizuho Yamato
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Yukinori Ikeda
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Hedele Zeng
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Jun-ichi Kawabe
- grid.252427.40000 0000 8638 2724Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-higashi, Asahikawa, Hokkaido 078-8510 Japan
| | - Yukiko T. Matsunaga
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| |
Collapse
|