101
|
Nguyen N, Thurgood P, Sekar NC, Chen S, Pirogova E, Peter K, Baratchi S, Khoshmanesh K. Microfluidic models of the human circulatory system: versatile platforms for exploring mechanobiology and disease modeling. Biophys Rev 2021; 13:769-786. [PMID: 34777617 DOI: 10.1007/s12551-021-00815-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human circulatory system is a marvelous fluidic system, which is very sensitive to biophysical and biochemical cues. The current animal and cell culture models do not recapitulate the functional properties of the human circulatory system, limiting our ability to fully understand the complex biological processes underlying the dysfunction of this multifaceted system. In this review, we discuss the unique ability of microfluidic systems to recapitulate the biophysical, biochemical, and functional properties of the human circulatory system. We also describe the remarkable capacity of microfluidic technologies for exploring the complex mechanobiology of the cardiovascular system, mechanistic studying of cardiovascular diseases, and screening cardiovascular drugs with the additional benefit of reducing the need for animal models. We also discuss opportunities for further advancement in this exciting field.
Collapse
Affiliation(s)
- Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Sheng Chen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiometabolic Health, The University of Melbourne, Parkville, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | | |
Collapse
|
102
|
M Abdelgawad L, Salah N, Sabry D, Abdelgwad M. Efficacy of Photobiomodulation and Vitamin D on Odontogenic Activity of Human Dental Pulp Stem Cells. J Lasers Med Sci 2021; 12:e30. [PMID: 34733753 DOI: 10.34172/jlms.2021.30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 09/05/2020] [Indexed: 11/09/2022]
Abstract
Introduction: The regeneration of dental pulp tissue using human dental pulp stem cells (HDPSCs) has attracted increasing attention in recent years. Recent studies have suggested that several factors such as photobiomodulation (PBM) and vitamin D affect the proliferation and differentiation of HDPSCs. Therefore, the present study evaluated the effects of PBM and vitamin D on odontogenic differentiation of HDPSCs for dentin -like tissue formation. Methods: HDPSCs were collected, isolated, and characterized and then divided into six groups: group I, control; group II, vitamin D (10-7 Mol); group III, irradiation at 1 J/cm2 of 810 nm diode laser; group IV, irradiation at 1 J/cm2 and culture with vitamin D; group V, irradiation at 2 J/cm2, and group VI, irradiation at 2 J/cm2 and culture with vitamin D, cell viability assay was measured through MTT. Alkaline phosphatase (ALP) enzyme activity and mRNA levels of vascular endothelial growth factor (VEGF), bone morphogenic protein-2 (BMP-2), and dentin sialophosphoprotein (DSPP) were also assessed. Results: PBM at 1 and 2 J/cm2 combined with vitamin D significantly promoted HDPSCs proliferation through MTT assay and odontogenic differentiation through gene expression of VEGF, BMP-2, and DSPP levels (P < 0.0001). Conclusion: PBM at 2 J/cm2 combined with vitamin D enhanced the HDPSCs proliferation and odontogenic differentiation and thus could be a novel strategy for dentin regeneration in dentistry.
Collapse
Affiliation(s)
- Latifa M Abdelgawad
- Professor of Medical Laser Applications Department, National Institute of Laser Enhanced Sciences (NILES), Cairo University, Cairo, Egypt
| | - Nehal Salah
- Medical Laser Applications Department, National Institute of Laser Enhanced Sciences (NILES), Cairo University, Cairo, Egypt
| | - Dina Sabry
- Professor of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Marwa Abdelgwad
- Lecturer of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
103
|
Pattanayak P, Singh SK, Gulati M, Vishwas S, Kapoor B, Chellappan DK, Anand K, Gupta G, Jha NK, Gupta PK, Prasher P, Dua K, Dureja H, Kumar D, Kumar V. Microfluidic chips: recent advances, critical strategies in design, applications and future perspectives. MICROFLUIDICS AND NANOFLUIDICS 2021; 25:99. [PMID: 34720789 PMCID: PMC8547131 DOI: 10.1007/s10404-021-02502-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/19/2021] [Indexed: 05/12/2023]
Abstract
Microfluidic chip technology is an emerging tool in the field of biomedical application. Microfluidic chip includes a set of groves or microchannels that are engraved on different materials (glass, silicon, or polymers such as polydimethylsiloxane or PDMS, polymethylmethacrylate or PMMA). The microchannels forming the microfluidic chip are interconnected with each other for desired results. This organization of microchannels trapped into the microfluidic chip is associated with the outside by inputs and outputs penetrating through the chip, as an interface between the macro- and miniature world. With the help of a pump and a chip, microfluidic chip helps to determine the behavioral change of the microfluids. Inside the chip, there are microfluidic channels that permit the processing of the fluid, for example, blending and physicochemical responses. Microfluidic chip has numerous points of interest including lesser time and reagent utilization and alongside this, it can execute numerous activities simultaneously. The miniatured size of the chip fastens the reaction as the surface area increases. It is utilized in different biomedical applications such as food safety sensing, peptide analysis, tissue engineering, medical diagnosis, DNA purification, PCR activity, pregnancy, and glucose estimation. In the present study, the design of various microfluidic chips has been discussed along with their biomedical applications.
Collapse
Affiliation(s)
- Prapti Pattanayak
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Dinesh Kumar Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein, South Africa
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310 India
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Plot no. 32-34, Knowledge Park III, Greater Noida, Uttar Pradesh 201310 India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007 India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007 Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana 12401 India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, 173229 India
| | - Vijay Kumar
- School of Bioengineering and Bioscience, Lovely Professional University, Phagwara, Punjab 144411 India
| |
Collapse
|
104
|
Recent Advances in Cardiac Tissue Engineering for the Management of Myocardium Infarction. Cells 2021; 10:cells10102538. [PMID: 34685518 PMCID: PMC8533887 DOI: 10.3390/cells10102538] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Myocardium Infarction (MI) is one of the foremost cardiovascular diseases (CVDs) causing death worldwide, and its case numbers are expected to continuously increase in the coming years. Pharmacological interventions have not been at the forefront in ameliorating MI-related morbidity and mortality. Stem cell-based tissue engineering approaches have been extensively explored for their regenerative potential in the infarcted myocardium. Recent studies on microfluidic devices employing stem cells under laboratory set-up have revealed meticulous events pertaining to the pathophysiology of MI occurring at the infarcted site. This discovery also underpins the appropriate conditions in the niche for differentiating stem cells into mature cardiomyocyte-like cells and leads to engineering of the scaffold via mimicking of native cardiac physiological conditions. However, the mode of stem cell-loaded engineered scaffolds delivered to the site of infarction is still a challenging mission, and yet to be translated to the clinical setting. In this review, we have elucidated the various strategies developed using a hydrogel-based system both as encapsulated stem cells and as biocompatible patches loaded with cells and applied at the site of infarction.
Collapse
|
105
|
Wnt Modulation Enhances Otic Differentiation by Facilitating the Enucleation Process but Develops Unnecessary Cardiac Structures. Int J Mol Sci 2021; 22:ijms221910306. [PMID: 34638648 PMCID: PMC8508801 DOI: 10.3390/ijms221910306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 01/08/2023] Open
Abstract
Otic organoids have the potential to resolve current challenges in hearing loss research. The reproduction of the delicate and complex structure of the mammalian cochlea using organoids requires high efficiency and specificity. Recent attempts to strengthen otic organoids have focused on the effects of the Wnt signaling pathway on stem cell differentiation. One important aspect of this is the evaluation of undesirable effects of differentiation after Wnt activation. In the present study, we differentiated mouse embryonic stem cell embryoid bodies (EB) into otic organoids and observed two morphologies with different cell fates. EBs that underwent a core ejection process, or ‘enucleation,’ were similar to previously reported inner ear organoids. Meanwhile, EBs that retained their core demonstrated features characteristic of neural organoids. The application of a Wnt agonist during the maturation phase increased enucleation, as well as otic organoid formation, in turn leading to sensory hair cell-like cell generation. However, with a longer incubation period, Wnt activation also led to EBs with ‘beating’ organoids that exhibited spontaneous movement. This observation emphasizes the necessity of optimizing Wnt enhancement for the differentiation of specific cells, such as those found in the inner ear.
Collapse
|
106
|
Prasad M, Kumar R, Buragohain L, Kumari A, Ghosh M. Organoid Technology: A Reliable Developmental Biology Tool for Organ-Specific Nanotoxicity Evaluation. Front Cell Dev Biol 2021; 9:696668. [PMID: 34631696 PMCID: PMC8495170 DOI: 10.3389/fcell.2021.696668] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
Engineered nanomaterials are bestowed with certain inherent physicochemical properties unlike their parent materials, rendering them suitable for the multifaceted needs of state-of-the-art biomedical, and pharmaceutical applications. The log-phase development of nano-science along with improved "bench to beside" conversion carries an enhanced probability of human exposure with numerous nanoparticles. Thus, toxicity assessment of these novel nanoscale materials holds a key to ensuring the safety aspects or else the global biome will certainly face a debacle. The toxicity may span from health hazards due to direct exposure to indirect means through food chain contamination or environmental pollution, even causing genotoxicity. Multiple ways of nanotoxicity evaluation include several in vitro and in vivo methods, with in vitro methods occupying the bulk of the "experimental space." The underlying reason may be multiple, but ethical constraints in in vivo animal experiments are a significant one. Two-dimensional (2D) monoculture is undoubtedly the most exploited in vitro method providing advantages in terms of cost-effectiveness, high throughput, and reproducibility. However, it often fails to mimic a tissue or organ which possesses a defined three-dimensional structure (3D) along with intercellular communication machinery. Instead, microtissues such as spheroids or organoids having a precise 3D architecture and proximate in vivo tissue-like behavior can provide a more realistic evaluation than 2D monocultures. Recent developments in microfluidics and bioreactor-based organoid synthesis have eased the difficulties to prosper nano-toxicological analysis in organoid models surpassing the obstacle of ethical issues. The present review will enlighten applications of organoids in nanotoxicological evaluation, their advantages, and prospects toward securing commonplace nano-interventions.
Collapse
Affiliation(s)
- Minakshi Prasad
- Department of Animal Biotechnology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Rajesh Kumar
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Lukumoni Buragohain
- Department of Animal Biotechnology, College of Veterinary Science, Assam Agricultural University, Guwahati, India
| | | | - Mayukh Ghosh
- Department of Veterinary Physiology and Biochemistry, RGSC, Banaras Hindu University, Varanasi, India
| |
Collapse
|
107
|
Isochoric supercooled preservation and revival of human cardiac microtissues. Commun Biol 2021; 4:1118. [PMID: 34552201 PMCID: PMC8458396 DOI: 10.1038/s42003-021-02650-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Low-temperature biopreservation and 3D tissue engineering present two differing routes towards eventual on-demand access to transplantable biologics, but recent advances in both fields present critical new opportunities for crossover between them. In this work, we demonstrate sub-zero centigrade preservation and revival of autonomously beating three-dimensional human induced pluripotent stem cell (hiPSC)-derived cardiac microtissues via isochoric supercooling, without the use of chemical cryoprotectants. We show that these tissues can cease autonomous beating during preservation and resume it after warming, that the supercooling process does not affect sarcomere structural integrity, and that the tissues maintain responsiveness to drug exposure following revival. Our work suggests both that functional three dimensional (3D) engineered tissues may provide an excellent high-content, low-risk testbed to study complex tissue biopreservation in a genetically human context, and that isochoric supercooling may provide a robust method for preserving and reviving engineered tissues themselves. Powell-Palm et al. demonstrate sub-zero centigrade preservation and revival of autonomously beating, 3D human induced pluripotent stem cell (hiPSC)-derived cardiac microtissues via isochoric supercooling, without the use of chemical cryoprotectants. Their study suggests that functional 3D engineered tissues may provide a high-content, low-risk testbed to study complex tissue biopreservation in a genetically human context, and that isochoric supercooling may provide a robust method for preserving and reviving engineered tissues themselves.
Collapse
|
108
|
Photobiomodulation therapy preconditioning modifies nitric oxide pathway and oxidative stress in human-induced pluripotent stem cell-derived ventricular cardiomyocytes treated with doxorubicin. Lasers Med Sci 2021; 37:1667-1675. [PMID: 34536182 DOI: 10.1007/s10103-021-03416-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic that exhibits high heart toxicity. Human-induced pluripotent stem cell-derived ventricular cardiomyocytes (hiPSC-vCMs) are important in vitro models for testing drug cardiotoxicity. Photobiomodulation therapy (PBMT) is a non-invasive therapy that stimulates cells growth and self-repair using light irradiation. This study aimed to investigate the in vitro effects of PBMT preconditioning on cardiotoxicity induced by DOX. HiPSC-vCMs were treated with PBMT for 500 s, followed by the addition of 2 μM DOX. LED irradiation preconditioning parameters were at 660 nm with an irradiance of 10 mW/cm2, performing 5 J/cm2, followed by 24-h DOX exposure (2 μM). Human iPSC-vCMs treated with 2 μM DOX or irradiated with PBMT composed the second and third groups, respectively. The control group did neither receive PBMT preconditioning nor DOX and was irradiated with a white standard lamp. Cells from all groups were collected to perform mRNA and miRNA expressions quantification. PBMT, when applied before the DOX challenge, restored the viability of hiPSC-vCMs and reduced ROS levels. Although downregulated by DOX, myocardial UCP2 mRNA expression presented marked upregulation after PBMT preconditioning. Expression of eNOS and UCP2 mRNA and NO production were decreased after DOX exposure, and PBMT preconditioning before the DOX challenge reversed these changes. Moreover, our data indicated that PBMT preconditioning lowered the miR-24 expression. Our data suggested that PBMT preconditioning ameliorated in vitro DOX-induced cardiotoxicity on transcription level, restoring NO levels and reducing oxidative stress.
Collapse
|
109
|
Electromechanical Stimulation of 3D Cardiac Microtissues in a Heart-on-Chip Model. Methods Mol Biol 2021; 2373:133-157. [PMID: 34520011 DOI: 10.1007/978-1-0716-1693-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Modeling human cardiac tissues in vitro is essential to elucidate the biological mechanisms related to the heart physiopathology, possibly paving the way for new treatments. Organs-on-chips have emerged as innovative tools able to recreate tissue-specific microenvironments, guiding the development of miniaturized models and offering the opportunity to directly analyze functional readouts. Here we describe the fabrication and operational procedures for the development of a heart-on-chip model, reproducing cardiac biomimetic microenvironment. The device provides 3D cardiac microtissue with a synchronized electromechanical stimulation to support the tissue development. We additionally describe procedures for characterizing tissue evolution and functionality through immunofluorescence, real time qPCR, calcium imaging and microtissue contractility investigations.
Collapse
|
110
|
Aboul-Soud MAM, Alzahrani AJ, Mahmoud A. Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies, Disease Modelling and Drug Screening. Cells 2021; 10:cells10092319. [PMID: 34571968 PMCID: PMC8467501 DOI: 10.3390/cells10092319] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has made an invaluable contribution to the field of regenerative medicine, paving way for identifying the true potential of human embryonic stem cells (ESCs). Since the controversy around ethicality of ESCs continue to be debated, iPSCs have been used to circumvent the process around destruction of the human embryo. The use of iPSCs have transformed biological research, wherein increasing number of studies are documenting nuclear reprogramming strategies to make them beneficial models for drug screening as well as disease modelling. The flexibility around the use of iPSCs include compatibility to non-invasive harvesting, and ability to source from patients with rare diseases. iPSCs have been widely used in cardiac disease modelling, studying inherited arrhythmias, neural disorders including Alzheimer’s disease, liver disease, and spinal cord injury. Extensive research around identifying factors that are involved in maintaining the identity of ESCs during induction of pluripotency in somatic cells is undertaken. The focus of the current review is to detail all the clinical translation research around iPSCs and the strength of its ever-growing potential in the clinical space.
Collapse
Affiliation(s)
- Mourad A. M. Aboul-Soud
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
- Correspondence:
| | - Alhusain J. Alzahrani
- Department of Clinical Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39524, Saudi Arabia;
| | - Amer Mahmoud
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| |
Collapse
|
111
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
112
|
Vunjak-Novakovic G, Ronaldson-Bouchard K, Radisic M. Organs-on-a-chip models for biological research. Cell 2021; 184:4597-4611. [PMID: 34478657 PMCID: PMC8417425 DOI: 10.1016/j.cell.2021.08.005] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
We explore the utility of bioengineered human tissues-individually or connected into physiological units-for biological research. While much smaller and simpler than their native counterparts, these tissues are complex enough to approximate distinct tissue phenotypes: molecular, structural, and functional. Unlike organoids, which form spontaneously and recapitulate development, "organs-on-a-chip" are engineered to display some specific functions of whole organs. Looking back, we discuss the key developments of this emerging technology. Thinking forward, we focus on the challenges faced to fully establish, validate, and utilize the fidelity of these models for biological research.
Collapse
|
113
|
Lu RXZ, Radisic M. Organ-on-a-chip platforms for evaluation of environmental nanoparticle toxicity. Bioact Mater 2021; 6:2801-2819. [PMID: 33665510 PMCID: PMC7900603 DOI: 10.1016/j.bioactmat.2021.01.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Despite showing a great promise in the field of nanomedicine, nanoparticles have gained a significant attention from regulatory agencies regarding their possible adverse health effects upon environmental exposure. Whether those nanoparticles are generated through intentional or unintentional means, the constant exposure to nanomaterials can inevitably lead to unintended consequences based on epidemiological data, yet the current understanding of nanotoxicity is insufficient relative to the rate of their emission in the environment and the lack of predictive platforms that mimic the human physiology. This calls for a development of more physiologically relevant models, which permit the comprehensive and systematic examination of toxic properties of nanoparticles. With the advancement in microfabrication techniques, scientists have shifted their focus on the development of an engineered system that acts as an intermediate between a well-plate system and animal models, known as organ-on-a-chips. The ability of organ-on-a-chip models to recapitulate in vivo like microenvironment and responses offers a new avenue for nanotoxicological research. In this review, we aim to provide overview of assessing potential risks of nanoparticle exposure using organ-on-a-chip systems and their potential to delineate biological mechanisms of epidemiological findings.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- The Heart and Stroke/Richard Lewar Centre of Excellence, Toronto, ON, Canada
| |
Collapse
|
114
|
Chu X, Wang M, Qiu X, Huang Y, Li T, Otieno E, Li N, Luo L, Xiao X. Strategies for constructing pluripotent stem cell- and progenitor cell-derived three-dimensional cardiac micro-tissues. J Biomed Mater Res A 2021; 110:488-503. [PMID: 34397148 DOI: 10.1002/jbm.a.37298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cardiac micro-tissue is a promising model for simulating the structural and functional features of heart in vitro. This scientific achievement provides a platform for exploration about the mechanisms on the development, damage, and regeneration of tissue, hence, paving a way toward development of novel therapies for heart diseases. However, 3D micro-tissue technology is still in its infant stages faced with many challenges such as incompleteness of the tissue microarchitecture, loss of the resident immune cells, poor reproducibility, and deficiencies in continuously feeding the nutrients and removing wastes during micro-tissue culturing. There is an urgent need to optimize the construction of 3D cardiac micro-tissue and improve functions of the involved cells. Therefore, scaffolds and cell resources for building 3D cardiac micro-tissues, strategies for inducing the maturation and functionalization of pluripotent stem cell- or cardiac progenitor cell-derived cardiomyocytes, and the major challenges were reviewed in this writing to enable future fabrication of 3D cardiac micro-tissues or organoids for drug screening, disease modeling, regeneration treatment, and so on.
Collapse
Affiliation(s)
- Xinyue Chu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mingyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China.,Institute of Laboratory Animals Science, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Husbandry Engineering, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yun Huang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Tong Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Edward Otieno
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Na Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Li Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
115
|
Charrez B, Charwat V, Siemons BA, Goswami I, Sakolish C, Luo YS, Finsberg H, Edwards AG, Miller EW, Rusyn I, Healy KE. Heart Muscle Microphysiological System for Cardiac Liability Prediction of Repurposed COVID-19 Therapeutics. Front Pharmacol 2021; 12:684252. [PMID: 34421592 PMCID: PMC8378272 DOI: 10.3389/fphar.2021.684252] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022] Open
Abstract
Despite global efforts, it took 7 months between the proclamation of global SARS-CoV-2 pandemic and the first FDA-approved treatment for COVID-19. During this timeframe, clinicians focused their efforts on repurposing drugs, such as hydroxychloroquine (HCQ) or azithromycin (AZM) to treat hospitalized COVID-19 patients. While clinical trials are time-consuming, the exponential increase in hospitalizations compelled the FDA to grant an emergency use authorization for HCQ and AZM as treatment for COVID-19, although there was limited evidence of their combined efficacy and safety. The authorization was revoked 4 months later, giving rise to controversial political and scientific debates illustrating important challenges such as premature authorization of potentially ineffective or unsafe therapeutics, while diverting resources from screening of effective drugs. Here we report on a preclinical drug screening platform, a cardiac microphysiological system (MPS), to rapidly identify clinically relevant cardiac liabilities associated with HCQ and AZM. The cardiac MPS is a microfabricated fluidic system in which cardiomyocytes derived from human induced pluripotent stem cells self-arrange into a uniaxially beating tissue. The drug response was measured using outputs that correlate with clinical measurements such as action potential duration (proxy for clinical QT interval) and drug-biomarker pairing. The cardiac MPS predicted clinical arrhythmias associated with QT prolongation and rhythm instabilities in tissues treated with HCQ. We found no change in QT interval upon acute exposure to AZM, while still observing a significant increase in arrhythmic events. These results suggest that this MPS can not only predict arrhythmias, but it can also identify arrhythmias even when QT prolongation is absent. When exposed to HCQ and AZM polytherapy, this MPS faithfully reflected clinical findings, in that the combination of drugs synergistically increased QT interval when compared to single drug exposure, while not worsening the overall frequency of arrhythmic events. The high content cardiac MPS can rapidly evaluate the cardiac safety of potential therapeutics, ultimately accelerating patients' access to safe and effective treatments.
Collapse
Affiliation(s)
- Bérénice Charrez
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, United States
| | - Verena Charwat
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, United States
| | - Brian A. Siemons
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, United States
| | - Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, United States
| | - Courtney Sakolish
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Yu-Syuan Luo
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | | | - Andrew G. Edwards
- Department of Pharmacology, School of Medicine, University of California at Davis, Davis, CA, United States
| | - Evan W. Miller
- Department of Chemistry, University of California at Berkeley, Berkeley, CA, United States
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA, United States
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Kevin E. Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, United States
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, CA, United States
| |
Collapse
|
116
|
Chen Y, Alba M, Tieu T, Tong Z, Minhas RS, Rudd D, Voelcker NH, Cifuentes-Rius A, Elnathan R. Engineering Micro–Nanomaterials for Biomedical Translation. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton VIC 3168 Australia
| | - Ziqiu Tong
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Rajpreet Singh Minhas
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - David Rudd
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
- Department of Materials Science and Engineering Monash University 22 Alliance Lane Clayton VIC 3168 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO) Clayton VIC 3168 Australia
- INM-Leibniz Institute for New Materials Campus D2 2 Saarbrücken 66123 Germany
| | - Anna Cifuentes-Rius
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences Monash University 381 Royal Parade Parkville VIC 3052 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton VIC 3168 Australia
- Department of Materials Science and Engineering Monash University 22 Alliance Lane Clayton VIC 3168 Australia
| |
Collapse
|
117
|
Monckton CP, Brown GE, Khetani SR. Latest impact of engineered human liver platforms on drug development. APL Bioeng 2021; 5:031506. [PMID: 34286173 PMCID: PMC8286174 DOI: 10.1063/5.0051765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition, which is partly due to differences between preclinical animals and humans in metabolic pathways. Therefore, in vitro human liver models are utilized in biopharmaceutical practice to mitigate DILI risk and assess related mechanisms of drug transport and metabolism. However, liver cells lose phenotypic functions within 1–3 days in two-dimensional monocultures on collagen-coated polystyrene/glass, which precludes their use to model the chronic effects of drugs and disease stimuli. To mitigate such a limitation, bioengineers have adapted tools from the semiconductor industry and additive manufacturing to precisely control the microenvironment of liver cells. Such tools have led to the fabrication of advanced two-dimensional and three-dimensional human liver platforms for different throughput needs and assay endpoints (e.g., micropatterned cocultures, spheroids, organoids, bioprinted tissues, and microfluidic devices); such platforms have significantly enhanced liver functions closer to physiologic levels and improved functional lifetime to >4 weeks, which has translated to higher sensitivity for predicting drug outcomes and enabling modeling of diseased phenotypes for novel drug discovery. Here, we focus on commercialized engineered liver platforms and case studies from the biopharmaceutical industry showcasing their impact on drug development. We also discuss emerging multi-organ microfluidic devices containing a liver compartment that allow modeling of inter-tissue crosstalk following drug exposure. Finally, we end with key requirements for engineered liver platforms to become routine fixtures in the biopharmaceutical industry toward reducing animal usage and providing patients with safe and efficacious drugs with unprecedented speed and reduced cost.
Collapse
Affiliation(s)
- Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
118
|
Liu Y, Sun L, Zhang H, Shang L, Zhao Y. Microfluidics for Drug Development: From Synthesis to Evaluation. Chem Rev 2021; 121:7468-7529. [PMID: 34024093 DOI: 10.1021/acs.chemrev.0c01289] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug development is a long process whose main content includes drug synthesis, drug delivery, and drug evaluation. Compared with conventional drug development procedures, microfluidics has emerged as a revolutionary technology in that it offers a miniaturized and highly controllable environment for bio(chemical) reactions to take place. It is also compatible with analytical strategies to implement integrated and high-throughput screening and evaluations. In this review, we provide a comprehensive summary of the entire microfluidics-based drug development system, from drug synthesis to drug evaluation. The challenges in the current status and the prospects for future development are also discussed. We believe that this review will promote communications throughout diversified scientific and engineering communities that will continue contributing to this burgeoning field.
Collapse
Affiliation(s)
- Yuxiao Liu
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyu Sun
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Hui Zhang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Luoran Shang
- Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
119
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
120
|
Andrysiak K, Stępniewski J, Dulak J. Human-induced pluripotent stem cell-derived cardiomyocytes, 3D cardiac structures, and heart-on-a-chip as tools for drug research. Pflugers Arch 2021; 473:1061-1085. [PMID: 33629131 PMCID: PMC8245367 DOI: 10.1007/s00424-021-02536-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
Development of new drugs is of high interest for the field of cardiac and cardiovascular diseases, which are a dominant cause of death worldwide. Before being allowed to be used and distributed, every new potentially therapeutic compound must be strictly validated during preclinical and clinical trials. The preclinical studies usually involve the in vitro and in vivo evaluation. Due to the increasing reporting of discrepancy in drug effects in animal and humans and the requirement to reduce the number of animals used in research, improvement of in vitro models based on human cells is indispensable. Primary cardiac cells are difficult to access and maintain in cell culture for extensive experiments; therefore, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) became an excellent alternative. This technology enables a production of high number of patient- and disease-specific cardiomyocytes and other cardiac cell types for a large-scale research. The drug effects can be extensively evaluated in the context of electrophysiological responses with a use of well-established tools, such as multielectrode array (MEA), patch clamp, or calcium ion oscillation measurements. Cardiotoxicity, which is a common reason for withdrawing drugs from marketing or rejection at final stages of clinical trials, can be easily verified with a use of hiPSC-CM model providing a prediction of human-specific responses and higher safety of clinical trials involving patient cohort. Abovementioned studies can be performed using two-dimensional cell culture providing a high-throughput and relatively lower costs. On the other hand, more complex structures, such as engineered heart tissue, organoids, or spheroids, frequently applied as co-culture systems, represent more physiological conditions and higher maturation rate of hiPSC-derived cells. Furthermore, heart-on-a-chip technology has recently become an increasingly popular tool, as it implements controllable culture conditions, application of various stimulations and continuous parameters read-out. This paper is an overview of possible use of cardiomyocytes and other cardiac cell types derived from hiPSC as in vitro models of heart in drug research area prepared on the basis of latest scientific reports and providing thorough discussion regarding their advantages and limitations.
Collapse
Affiliation(s)
- Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
121
|
Zhao Q, Cole T, Zhang Y, Tang SY. Mechanical Strain-Enabled Reconstitution of Dynamic Environment in Organ-on-a-Chip Platforms: A Review. MICROMACHINES 2021; 12:765. [PMID: 34203533 PMCID: PMC8304354 DOI: 10.3390/mi12070765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
Organ-on-a-chip (OOC) uses the microfluidic 3D cell culture principle to reproduce organ- or tissue-level functionality at a small scale instead of replicating the entire human organ. This provides an alternative to animal models for drug development and environmental toxicology screening. In addition to the biomimetic 3D microarchitecture and cell-cell interactions, it has been demonstrated that mechanical stimuli such as shear stress and mechanical strain significantly influence cell behavior and their response to pharmaceuticals. Microfluidics is capable of precisely manipulating the fluid of a microenvironment within a 3D cell culture platform. As a result, many OOC prototypes leverage microfluidic technology to reproduce the mechanically dynamic microenvironment on-chip and achieve enhanced in vitro functional organ models. Unlike shear stress that can be readily generated and precisely controlled using commercial pumping systems, dynamic systems for generating proper levels of mechanical strains are more complicated, and often require miniaturization and specialized designs. As such, this review proposes to summarize innovative microfluidic OOC platforms utilizing mechanical actuators that induce deflection of cultured cells/tissues for replicating the dynamic microenvironment of human organs.
Collapse
Affiliation(s)
- Qianbin Zhao
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Tim Cole
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (T.C.); (Y.Z.)
| | - Yuxin Zhang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (T.C.); (Y.Z.)
| | - Shi-Yang Tang
- Department of Electronic, Electrical and Systems Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (T.C.); (Y.Z.)
| |
Collapse
|
122
|
Tavakol DN, Fleischer S, Vunjak-Novakovic G. Harnessing organs-on-a-chip to model tissue regeneration. Cell Stem Cell 2021; 28:993-1015. [PMID: 34087161 DOI: 10.1016/j.stem.2021.05.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue engineering has markedly matured since its early beginnings in the 1980s. In addition to the original goal to regenerate damaged organs, the field has started to explore modeling of human physiology "in a dish." Induced pluripotent stem cell (iPSC) technologies now enable studies of organ regeneration and disease modeling in a patient-specific context. We discuss the potential of "organ-on-a-chip" systems to study regenerative therapies with focus on three distinct organ systems: cardiac, respiratory, and hematopoietic. We propose that the combinatorial studies of human tissues at these two scales would help realize the translational potential of tissue engineering.
Collapse
Affiliation(s)
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Medicine, Columbia University, New York, NY.
| |
Collapse
|
123
|
Cruz-Moreira D, Visone R, Vasques-Nóvoa F, S Barros A, Leite-Moreira A, Redaelli A, Moretti M, Rasponi M. Assessing the influence of perfusion on cardiac microtissue maturation: A heart-on-chip platform embedding peristaltic pump capabilities. Biotechnol Bioeng 2021; 118:3128-3137. [PMID: 34019719 PMCID: PMC8362142 DOI: 10.1002/bit.27836] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 01/24/2023]
Abstract
Heart‐on‐chip is an unprecedented technology for recapitulating key biochemical and biophysical cues in cardiac pathophysiology. Several designs have been proposed to improve its ability to mimic the native tissue and establish it as a reliable research platform. However, despite mimicking one of most vascularized organs, reliable strategies to deliver oxygen and substrates to densely packed constructs of metabolically demanding cells remain unsettled. Herein, we describe a new heart‐on‐chip platform with precise fluid control, integrating an on‐chip peristaltic pump, allowing automated and fine control over flow on channels flanking a 3D cardiac culture. The application of distinct flow rates impacted on temporal dynamics of microtissue structural and transcriptional maturation, improving functional performance. Moreover, a widespread transcriptional response was observed, suggesting flow‐mediated activation of critical pathways of cardiomyocyte structural and functional maturation and inhibition of cardiomyocyte hypoxic injury. In conclusion, the present design represents an important advance in bringing engineered cardiac microtissues closer to the native heart, overcoming traditional bulky off‐chip fluid handling systems, improving microtissue performance, and matching oxygen and energy substrate requirements of metabolically active constructs, avoiding cellular hypoxia. Distinct flow patterns differently impact on microtissue performance and gene expression program.
Collapse
Affiliation(s)
- Daniela Cruz-Moreira
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Roberta Visone
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Francisco Vasques-Nóvoa
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - António S Barros
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Adelino Leite-Moreira
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| |
Collapse
|
124
|
Zheng F, Xiao Y, Liu H, Fan Y, Dao M. Patient-Specific Organoid and Organ-on-a-Chip: 3D Cell-Culture Meets 3D Printing and Numerical Simulation. Adv Biol (Weinh) 2021; 5:e2000024. [PMID: 33856745 PMCID: PMC8243895 DOI: 10.1002/adbi.202000024] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/13/2021] [Indexed: 12/11/2022]
Abstract
The last few decades have witnessed diversified in vitro models to recapitulate the architecture and function of living organs or tissues and contribute immensely to advances in life science. Two novel 3D cell culture models: 1) Organoid, promoted mainly by the developments of stem cell biology and 2) Organ-on-a-chip, enhanced primarily due to microfluidic technology, have emerged as two promising approaches to advance the understanding of basic biological principles and clinical treatments. This review describes the comparable distinct differences between these two models and provides more insights into their complementarity and integration to recognize their merits and limitations for applicable fields. The convergence of the two approaches to produce multi-organoid-on-a-chip or human organoid-on-a-chip is emerging as a new approach for building 3D models with higher physiological relevance. Furthermore, rapid advancements in 3D printing and numerical simulations, which facilitate the design, manufacture, and results-translation of 3D cell culture models, can also serve as novel tools to promote the development and propagation of organoid and organ-on-a-chip systems. Current technological challenges and limitations, as well as expert recommendations and future solutions to address the promising combinations by incorporating organoids, organ-on-a-chip, 3D printing, and numerical simulation, are also summarized.
Collapse
Affiliation(s)
- Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Yuminghao Xiao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
125
|
Benlian BR, Klier PEZ, Martinez KN, Schwinn MK, Kirkland TA, Miller EW. Small Molecule-Protein Hybrid for Voltage Imaging via Quenching of Bioluminescence. ACS Sens 2021; 6:1857-1863. [PMID: 33723996 DOI: 10.1021/acssensors.1c00058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We report a small-molecule enzyme pair for optical voltage sensing via quenching of bioluminescence. This quenching bioluminescent voltage indicator, or Q-BOLT, pairs the dark absorbing, voltage-sensitive dipicrylamine with membrane-localized bioluminescence from the luciferase NanoLuc (NLuc). As a result, bioluminescence is quenched through resonance energy transfer (QRET) as a function of membrane potential. Fusion of HaloTag to NLuc creates a two-acceptor bioluminescence resonance energy transfer (BRET) system when a tetramethylrhodamine (TMR) HaloTag ligand is ligated to HaloTag. In this mode, Q-BOLT is capable of providing direct visualization of changes in membrane potential in live cells via three distinct readouts: change in QRET, BRET, and the ratio between bioluminescence emission and BRET. Q-BOLT can provide up to a 29% change in bioluminescence (ΔBL/BL) and >100% ΔBRET/BRET per 100 mV change in HEK 293T cells, without the need for excitation light. In cardiac monolayers derived from human-induced pluripotent stem cells (hiPSCs), Q-BOLT readily reports on membrane potential oscillations. Q-BOLT is the first example of a hybrid small molecule-protein voltage indicator that does not require excitation light and may be useful in contexts where excitation light is limiting.
Collapse
Affiliation(s)
| | | | | | | | - Thomas A. Kirkland
- Promega Biosciences LLC, San Luis Obispo, California 93401, United States
| | | |
Collapse
|
126
|
A Paradigm Shift in Tissue Engineering: From a Top–Down to a Bottom–Up Strategy. Processes (Basel) 2021. [DOI: 10.3390/pr9060935] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Tissue engineering (TE) was initially designed to tackle clinical organ shortage problems. Although some engineered tissues have been successfully used for non-clinical applications, very few (e.g., reconstructed human skin) have been used for clinical purposes. As the current TE approach has not achieved much success regarding more broad and general clinical applications, organ shortage still remains a challenging issue. This very limited clinical application of TE can be attributed to the constraints in manufacturing fully functional tissues via the traditional top–down approach, where very limited cell types are seeded and cultured in scaffolds with equivalent sizes and morphologies as the target tissues. The newly proposed developmental engineering (DE) strategy towards the manufacture of fully functional tissues utilises a bottom–up approach to mimic developmental biology processes by implementing gradual tissue assembly alongside the growth of multiple cell types in modular scaffolds. This approach may overcome the constraints of the traditional top–down strategy as it can imitate in vivo-like tissue development processes. However, several essential issues must be considered, and more mechanistic insights of the fundamental, underpinning biological processes, such as cell–cell and cell–material interactions, are necessary. The aim of this review is to firstly introduce and compare the number of cell types, the size and morphology of the scaffolds, and the generic tissue reconstruction procedures utilised in the top–down and the bottom–up strategies; then, it will analyse their advantages, disadvantages, and challenges; and finally, it will briefly discuss the possible technologies that may overcome some of the inherent limitations of the bottom–up strategy.
Collapse
|
127
|
Schneider S, Gruner D, Richter A, Loskill P. Membrane integration into PDMS-free microfluidic platforms for organ-on-chip and analytical chemistry applications. LAB ON A CHIP 2021; 21:1866-1885. [PMID: 33949565 DOI: 10.1039/d1lc00188d] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Membranes play a crucial role in many microfluidic systems, enabling versatile applications in highly diverse research fields. However, the tight and robust integration of membranes into microfluidic systems requires complex fabrication processes. Most integration approaches, so far, rely on polydimethylsiloxane (PDMS) as base material for the microfluidic chips. Several limitations of PDMS have resulted in the transition of many microfluidic approaches to PDMS-free systems using alternative materials such as thermoplastics. To integrate membranes in those PDMS-free systems, novel alternative approaches are required. This review provides an introduction into microfluidic systems applying membrane technology for analytical systems and organ-on-chip as well as a comprehensive overview of methods for the integration of membranes into PDMS-free systems. The overview and examples will provide a valuable resource and starting point for any researcher that is aiming at implementing membranes in microfluidic systems without using PDMS.
Collapse
Affiliation(s)
- Stefan Schneider
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Denise Gruner
- Institut für Halbleiter- und Mikrosystemtechnik, Technische Universität Dresden, 01062 Dresden, Germany and Universitätsklinikum Carl Gustav Carus Dresden, Institut für Klinische Chemie und Laboratoriumsmedizin, 01307 Dresden, Germany
| | - Andreas Richter
- Institut für Halbleiter- und Mikrosystemtechnik, Technische Universität Dresden, 01062 Dresden, Germany
| | - Peter Loskill
- Department of Biomedical Science, Faculty of Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany. and NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| |
Collapse
|
128
|
Seguret M, Vermersch E, Jouve C, Hulot JS. Cardiac Organoids to Model and Heal Heart Failure and Cardiomyopathies. Biomedicines 2021; 9:563. [PMID: 34069816 PMCID: PMC8157277 DOI: 10.3390/biomedicines9050563] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiac tissue engineering aims at creating contractile structures that can optimally reproduce the features of human cardiac tissue. These constructs are becoming valuable tools to model some of the cardiac functions, to set preclinical platforms for drug testing, or to alternatively be used as therapies for cardiac repair approaches. Most of the recent developments in cardiac tissue engineering have been made possible by important advances regarding the efficient generation of cardiac cells from pluripotent stem cells and the use of novel biomaterials and microfabrication methods. Different combinations of cells, biomaterials, scaffolds, and geometries are however possible, which results in different types of structures with gradual complexities and abilities to mimic the native cardiac tissue. Here, we intend to cover key aspects of tissue engineering applied to cardiology and the consequent development of cardiac organoids. This review presents various facets of the construction of human cardiac 3D constructs, from the choice of the components to their patterning, the final geometry of generated tissues, and the subsequent readouts and applications to model and treat cardiac diseases.
Collapse
Affiliation(s)
- Magali Seguret
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Eva Vermersch
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Charlène Jouve
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Jean-Sébastien Hulot
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
- CIC1418 and DMU CARTE, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| |
Collapse
|
129
|
Lee-Montiel FT, Laemmle A, Charwat V, Dumont L, Lee CS, Huebsch N, Okochi H, Hancock MJ, Siemons B, Boggess SC, Goswami I, Miller EW, Willenbring H, Healy KE. Integrated Isogenic Human Induced Pluripotent Stem Cell-Based Liver and Heart Microphysiological Systems Predict Unsafe Drug-Drug Interaction. Front Pharmacol 2021; 12:667010. [PMID: 34025426 PMCID: PMC8138446 DOI: 10.3389/fphar.2021.667010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional (3D) microphysiological systems (MPSs) mimicking human organ function in vitro are an emerging alternative to conventional monolayer cell culture and animal models for drug development. Human induced pluripotent stem cells (hiPSCs) have the potential to capture the diversity of human genetics and provide an unlimited supply of cells. Combining hiPSCs with microfluidics technology in MPSs offers new perspectives for drug development. Here, the integration of a newly developed liver MPS with a cardiac MPS—both created with the same hiPSC line—to study drug–drug interaction (DDI) is reported. As a prominent example of clinically relevant DDI, the interaction of the arrhythmogenic gastroprokinetic cisapride with the fungicide ketoconazole was investigated. As seen in patients, metabolic conversion of cisapride to non-arrhythmogenic norcisapride in the liver MPS by the cytochrome P450 enzyme CYP3A4 was inhibited by ketoconazole, leading to arrhythmia in the cardiac MPS. These results establish integration of hiPSC-based liver and cardiac MPSs to facilitate screening for DDI, and thus drug efficacy and toxicity, isogenic in the same genetic background.
Collapse
Affiliation(s)
- Felipe T Lee-Montiel
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Alexander Laemmle
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States.,Institute of Clinical Chemistry and Department of Pediatrics, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Verena Charwat
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Laure Dumont
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States
| | - Caleb S Lee
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Nathaniel Huebsch
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, United States
| | | | - Brian Siemons
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Steven C Boggess
- Department of Chemistry, University of California Berkeley, Berkeley, CA, United States
| | - Ishan Goswami
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| | - Evan W Miller
- Departments of Chemistry and Molecular & Cell Biology, and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, United States
| | - Holger Willenbring
- Department of Surgery, Division of Transplant Surgery, Liver Center and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States
| | - Kevin E Healy
- Departments of Bioengineering, and Materials Science & Engineering, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
130
|
Jayne RK, Karakan MÇ, Zhang K, Pierce N, Michas C, Bishop DJ, Chen CS, Ekinci KL, White AE. Direct laser writing for cardiac tissue engineering: a microfluidic heart on a chip with integrated transducers. LAB ON A CHIP 2021; 21:1724-1737. [PMID: 33949395 DOI: 10.1039/d0lc01078b] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
We have developed a microfluidic platform for engineering cardiac microtissues in highly-controlled microenvironments. The platform is fabricated using direct laser writing (DLW) lithography and soft lithography, and contains four separate devices. Each individual device houses a cardiac microtissue and is equipped with an integrated strain actuator and a force sensor. Application of external pressure waves to the platform results in controllable time-dependent forces on the microtissues. Conversely, oscillatory forces generated by the microtissues are transduced into measurable electrical outputs. We demonstrate the capabilities of this platform by studying the response of cardiac microtissues derived from human induced pluripotent stem cells (hiPSC) under prescribed mechanical loading and pacing. This platform will be used for fundamental studies and drug screening on cardiac microtissues.
Collapse
Affiliation(s)
- Rachael K Jayne
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA
| | - M Çağatay Karakan
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Noelle Pierce
- Photonics Center, Boston University, Boston, MA 02215, USA
| | - Christos Michas
- Photonics Center, Boston University, Boston, MA 02215, USA and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - David J Bishop
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA and Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA and Department of Physics, Boston University, Boston, MA 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Kamil L Ekinci
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA and Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA
| | - Alice E White
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA. and Photonics Center, Boston University, Boston, MA 02215, USA and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA and Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA and Department of Physics, Boston University, Boston, MA 02215, USA
| |
Collapse
|
131
|
Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discov 2021; 20:345-361. [PMID: 32913334 DOI: 10.1038/s41573-020-0079-3] [Citation(s) in RCA: 477] [Impact Index Per Article: 119.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
Organs-on-chips (OoCs), also known as microphysiological systems or 'tissue chips' (the terms are synonymous), have attracted substantial interest in recent years owing to their potential to be informative at multiple stages of the drug discovery and development process. These innovative devices could provide insights into normal human organ function and disease pathophysiology, as well as more accurately predict the safety and efficacy of investigational drugs in humans. Therefore, they are likely to become useful additions to traditional preclinical cell culture methods and in vivo animal studies in the near term, and in some cases replacements for them in the longer term. In the past decade, the OoC field has seen dramatic advances in the sophistication of biology and engineering, in the demonstration of physiological relevance and in the range of applications. These advances have also revealed new challenges and opportunities, and expertise from multiple biomedical and engineering fields will be needed to fully realize the promise of OoCs for fundamental and translational applications. This Review provides a snapshot of this fast-evolving technology, discusses current applications and caveats for their implementation, and offers suggestions for directions in the next decade.
Collapse
Affiliation(s)
- Lucie A Low
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| | - Christine Mummery
- Leiden University Medical Center, Leiden, Netherlands
- University of Twente, Enschede, Netherlands
| | - Brian R Berridge
- National Institute for Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Danilo A Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
132
|
Charrez B, Charwat V, Siemons B, Finsberg H, Miller EW, Edwards AG, Healy KE. In vitro safety "clinical trial" of the cardiac liability of drug polytherapy. Clin Transl Sci 2021; 14:1155-1165. [PMID: 33786981 PMCID: PMC8212738 DOI: 10.1111/cts.13038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Only a handful of US Food and Drug Administration (FDA) Emergency Use Authorizations exist for drug and biologic therapeutics that treat severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection. Potential therapeutics include repurposed drugs, some with cardiac liabilities. We report on a chronic preclinical drug screening platform, a cardiac microphysiological system (MPS), to assess cardiotoxicity associated with repurposed hydroxychloroquine (HCQ) and azithromycin (AZM) polytherapy in a mock phase I safety clinical trial. The MPS contained human heart muscle derived from induced pluripotent stem cells. The effect of drug response was measured using outputs that correlate with clinical measurements, such as QT interval (action potential duration) and drug-biomarker pairing. Chronic exposure (10 days) of heart muscle to HCQ alone elicited early afterdepolarizations and increased QT interval past 5 days. AZM alone elicited an increase in QT interval from day 7 onward, and arrhythmias were observed at days 8 and 10. Monotherapy results mimicked clinical trial outcomes. Upon chronic exposure to HCQ and AZM polytherapy, we observed an increase in QT interval on days 4-8. Interestingly, a decrease in arrhythmias and instabilities was observed in polytherapy relative to monotherapy, in concordance with published clinical trials. Biomarkers, most of them measurable in patients' serum, were identified for negative effects of monotherapy or polytherapy on tissue contractile function, morphology, and antioxidant protection. The cardiac MPS correctly predicted clinical arrhythmias associated with QT prolongation and rhythm instabilities. This high content system can help clinicians design their trials, rapidly project cardiac outcomes, and define new monitoring biomarkers to accelerate access of patients to safe coronavirus disease 2019 (COVID-19) therapeutics.
Collapse
Affiliation(s)
- Bérénice Charrez
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
| | - Verena Charwat
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
| | - Brian Siemons
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
| | | | - Evan W. Miller
- Department of ChemistryUniversity of CaliforniaBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Department of Molecular and Cell BiologyUniversity of California at BerkeleyBerkeleyCaliforniaUSA
| | - Andrew G. Edwards
- Department of PharmacologySchool of MedicineUniversity of California at DavisDavisCaliforniaUSA
| | - Kevin E. Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3)University of California at BerkeleyBerkeleyCaliforniaUSA
- Department of Materials Science and EngineeringUniversity of California at BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
133
|
Hargrove-Grimes P, Low LA, Tagle DA. Microphysiological systems: What it takes for community adoption. Exp Biol Med (Maywood) 2021; 246:1435-1446. [PMID: 33899539 DOI: 10.1177/15353702211008872] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microphysiological systems (MPS) are promising in vitro tools which could substantially improve the drug development process, particularly for underserved patient populations such as those with rare diseases, neural disorders, and diseases impacting pediatric populations. Currently, one of the major goals of the National Institutes of Health MPS program, led by the National Center for Advancing Translational Sciences (NCATS), is to demonstrate the utility of this emerging technology and help support the path to community adoption. However, community adoption of MPS technology has been hindered by a variety of factors including biological and technological challenges in device creation, issues with validation and standardization of MPS technology, and potential complications related to commercialization. In this brief Minireview, we offer an NCATS perspective on what current barriers exist to MPS adoption and provide an outlook on the future path to adoption of these in vitro tools.
Collapse
Affiliation(s)
- Passley Hargrove-Grimes
- 390834National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lucie A Low
- 390834National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danilo A Tagle
- 390834National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
134
|
Circulating high-sensitivity troponin T and microRNAs as markers of myocardial damage during childhood leukaemia treatment. Pediatr Res 2021; 89:1245-1252. [PMID: 32634817 DOI: 10.1038/s41390-020-1049-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/23/2020] [Accepted: 06/23/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND We investigated whether plasma high-sensitivity cardiac troponin T (hs-cTnT) and circulating heart-associated microRNA (miRs) are increased in children with leukaemias during anthracycline-based chemotherapeutic treatment. METHODS In vitro human pluripotent stem cell (hPSC)-derived cardiomyocyte model showed that miR-1, miR-133a, miR-208a, miR-208b, and miR-499 are released from cells into culture medium in a time- and dose-dependent manner on doxorubicin exposure. Left ventricular (LV) myocardial deformation and circulating heart-associated miRs and plasma hs-cTnT during and after completion of chemotherapy were determined in 40 children with newly diagnosed acute leukaemia. RESULTS Significant reduction of LV global longitudinal strain and strain rates were found within 1 week after completion of anthracycline therapy in the induction phase of treatment (all p < 0.05). Hs-cTnT level peaked and miR-1 increased significantly at this time point. Log-transformed hs-cTnT correlated negatively with LV global systolic longitudinal strain (r = -0.38, p < 0.001). Receiver operating characteristic analysis revealed that area under the curve for changes in plasma hs-cTnT from baseline and plasma miR-1 levels in detecting a reduction in ≥20% of global longitudinal strain were respectively 0.62 (95% CI 0.38-0.87) and 0.62 (95% CI 0.40-0.84). CONCLUSION Plasma hs-cTnT and circulating miR-1 may be useful markers of myocardial damage during chemotherapy in children with leukaemias. IMPACT Heart-associated miRNAs including miR-1, miR-133a, miR-208a, miR-208b,and miR-499 were increased in the culture medium upon exposure of hPSC-derived cardiomyocytes to doxorubicin. Only miR-1 increased significantly during anthracycline-based therapy in paediatric leukaemic patients. In paediatric leukaemic patients, plasma hs-cTnT and circulating level of miR-1 showed the most significant increase within 1 week after completion of anthracycline therapy in the induction treatment phase. The study provides the first evidence of progressive increase in circulating miR-1 and plasma hs-cTnT levels during the course of anthracycline-based therapy in children with leukaemias, with hs-cTnT level also associated with changes in LV myocardial deformation.
Collapse
|
135
|
Zubareva EV, Nadezhdin SV, Nadezhdina NA, Belyaeva VS, Burda YE, Avtina TV, Gudyrev OS, Kolesnik IM, Kulikova SY, Mishenin MO. 3D organotypic cell structures for drug development and Microorganism-Host interaction research. RESEARCH RESULTS IN PHARMACOLOGY 2021. [DOI: 10.3897/rrpharmacology.7.62118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: The article describes a new method of tissue engineering, which is based on the use of three-dimensional multicellular constructs consisting of stem cells that mimic the native tissue in vivo – organoids.
3D cell cultures: The currently existing model systems of three-dimensional cultures are described.
Characteristics of organoids and strategies for their culturing: The main approaches to the fabrication of 3D cell constructs using pluripotent (embryonic and induced) stem cells or adult stem cells are described.
Brain organoids (Cerebral organoids): Organoids of the brain, which are used to study the development of the human brain, are characterized, with the description of biology of generating region-specific cerebral organoids.
Lung organoids: Approaches to the generation of lung organoids are described, by means of pluripotent stem cells and lung tissue cell lines.
Liver organoids: The features of differentiation of stem cells into hepatocyte-like cells and the creation of 3D hepatic organoids are characterized.
Intestinal organoids: The formation of small intestine organoids from stem cells is described.
Osteochondral organoids: Fabrication of osteochondral organoids is characterised.
Use of organoids as test systems for drugs screening: The information on drug screening using organoids is provided.
Using organoids to model infectious diseases and study adaptive responses of microorganisms when interacting with the host: The use of organoids for modeling infectious diseases and studying the adaptive responses of microorganisms when interacting with the host organism is described.
Conclusion: The creation of three-dimensional cell structures that reproduce the structural and functional characteristics of tissue in vivo, makes it possible to study the biology of the body’s development, the features of intercellular interactions, screening drugs and co-cultivating with viruses, bacteria and parasites.
Collapse
|
136
|
Inbody SC, Sinquefield BE, Lewis JP, Horton RE. Biomimetic microsystems for cardiovascular studies. Am J Physiol Cell Physiol 2021; 320:C850-C872. [PMID: 33760660 DOI: 10.1152/ajpcell.00026.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional tissue culture platforms have been around for several decades and have enabled key findings in the cardiovascular field. However, these platforms failed to recreate the mechanical and dynamic features found within the body. Organs-on-chips (OOCs) are cellularized microfluidic-based devices that can mimic the basic structure, function, and responses of organs. These systems have been successfully utilized in disease, development, and drug studies. OOCs are designed to recapitulate the mechanical, electrical, chemical, and structural features of the in vivo microenvironment. Here, we review cardiovascular-themed OOC studies, design considerations, and techniques used to generate these cellularized devices. Furthermore, we will highlight the advantages of OOC models over traditional cell culture vessels, discuss implementation challenges, and provide perspectives on the state of the field.
Collapse
Affiliation(s)
- Shelby C Inbody
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Bridgett E Sinquefield
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Joshua P Lewis
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Renita E Horton
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
137
|
Song Y, Uchida H, Sharipol A, Piraino L, Mereness JA, Ingalls MH, Rebhahn J, Newlands SD, DeLouise LA, Ovitt CE, Benoit DSW. Development of a functional salivary gland tissue chip with potential for high-content drug screening. Commun Biol 2021; 4:361. [PMID: 33742114 PMCID: PMC7979686 DOI: 10.1038/s42003-021-01876-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 02/17/2021] [Indexed: 01/31/2023] Open
Abstract
Radiation therapy for head and neck cancers causes salivary gland dysfunction leading to permanent xerostomia. Limited progress in the discovery of new therapeutic strategies is attributed to the lack of in vitro models that mimic salivary gland function and allow high-throughput drug screening. We address this limitation by combining engineered extracellular matrices with microbubble (MB) array technology to develop functional tissue mimetics for mouse and human salivary glands. We demonstrate that mouse and human salivary tissues encapsulated within matrix metalloproteinase-degradable poly(ethylene glycol) hydrogels formed in MB arrays are viable, express key salivary gland markers, and exhibit polarized localization of functional proteins. The salivary gland mimetics (SGm) respond to calcium signaling agonists and secrete salivary proteins. SGm were then used to evaluate radiosensitivity and mitigation of radiation damage using a radioprotective compound. Altogether, SGm exhibit phenotypic and functional parameters of salivary glands, and provide an enabling technology for high-content/throughput drug testing.
Collapse
Affiliation(s)
- Yuanhui Song
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Hitoshi Uchida
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Azmeer Sharipol
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Lindsay Piraino
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jared A Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew H Ingalls
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Jonathan Rebhahn
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Shawn D Newlands
- Department of Otolaryngology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Lisa A DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, USA
- Materials Science Program, University of Rochester, Rochester, NY, USA
| | - Catherine E Ovitt
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Materials Science Program, University of Rochester, Rochester, NY, USA.
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
138
|
Tsui JH, Leonard A, Camp ND, Long JT, Nawas ZY, Chavanachat R, Smith AST, Choi JS, Dong Z, Ahn EH, Wolf-Yadlin A, Murry CE, Sniadecki NJ, Kim DH. Tunable electroconductive decellularized extracellular matrix hydrogels for engineering human cardiac microphysiological systems. Biomaterials 2021; 272:120764. [PMID: 33798964 DOI: 10.1016/j.biomaterials.2021.120764] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) offer tremendous potential when used to engineer human tissues for drug screening and disease modeling; however, phenotypic immaturity reduces assay reliability when translating in vitro results to clinical studies. To address this, we have developed hybrid hydrogels comprised of decellularized porcine myocardial extracellular matrix (dECM) and reduced graphene oxide (rGO) to provide a more instructive microenvironment for proper cell and tissue development. A tissue-specific protein profile was preserved post-decellularization, and through the modulation of rGO content and degree of reduction, the mechanical and electrical properties of the hydrogels could be tuned. Engineered heart tissues (EHTs) generated using dECM-rGO hydrogel scaffolds and hiPSC-derived cardiomyocytes exhibited significantly increased twitch forces and had increased expression of genes that regulate contractile function. Improvements in various aspects of electrophysiological function, such as calcium-handling, action potential duration, and conduction velocity, were also induced by the hybrid biomaterial. dECM-rGO hydrogels could also be used as a bioink to print cardiac tissues in a high-throughput manner, and these tissues were utilized to assess the proarrhythmic potential of cisapride. Action potential prolongation and beat interval irregularities was observed in dECM-rGO tissues at clinical doses of cisapride, indicating that the enhanced electrophysiological function of these tissues corresponded well with a capability to produce physiologically relevant drug responses.
Collapse
Affiliation(s)
- Jonathan H Tsui
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Andrea Leonard
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98105, USA
| | - Nathan D Camp
- Department of Genome Sciences, University of Washington, Seattle, WA, 98105, USA
| | - Joseph T Long
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA
| | - Zeid Y Nawas
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA
| | | | - Alec S T Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98105, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Jong Seob Choi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhipeng Dong
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Charles E Murry
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA; Department of Pathology, University of Washington, Seattle, WA, 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98105, USA; Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA; Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
139
|
Hnatiuk AP, Briganti F, Staudt DW, Mercola M. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 2021; 28:271-282. [PMID: 33740432 PMCID: PMC8054828 DOI: 10.1016/j.chembiol.2021.02.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/26/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) have emerged as a promising platform for pharmacogenomics and drug development. In cardiology, they make it possible to produce unlimited numbers of patient-specific human cells that reproduce hallmark features of heart disease in the culture dish. Their potential applications include the discovery of mechanism-specific therapeutics, the evaluation of safety and efficacy in a human context before a drug candidate reaches patients, and the stratification of patients for clinical trials. Although this new technology has the potential to revolutionize drug discovery, translational hurdles have hindered its widespread adoption for pharmaceutical development. Here we discuss recent progress in overcoming these hurdles that should facilitate the use of hiPSCs to develop new medicines and individualize therapies for heart disease.
Collapse
Affiliation(s)
- Anna P Hnatiuk
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Francesca Briganti
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David W Staudt
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
140
|
Landi M, Everitt J, Berridge B. Bioethical, Reproducibility, and Translational Challenges of Animal Models. ILAR J 2021; 62:60-65. [PMID: 33693624 DOI: 10.1093/ilar/ilaa027] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 07/11/2020] [Accepted: 09/09/2020] [Indexed: 01/17/2023] Open
Abstract
There is no prescribed stage or standardized point at which an animal model protocol is reviewed for reproducibility and translatability. The method of review for a reproducible and translatable study is not consistently documented in peer literature, and this is a major challenge for those working with animal models of human diseases. If the study is ill designed, it is impossible to perform an accurate harm/benefit analysis. In addition, there may be an ethical challenge if the work is not reproducible and translatable. Animal welfare regulations and other documents of control clearly state the role of the Institutional Animal Care and Use Committees are to look at science justification within the context of animal welfare. This article, concentrating on models not governed by regulations, outlines issues and offers recommendations for refining animal model review with a goal to improve study reproducibility and translatability.
Collapse
Affiliation(s)
- Margaret Landi
- GSK Pharmaceuticals, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Jeffrey Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - B Berridge
- National Institute of Environmental Health Sciences, 111 T. W. Alexander Dr. Research Triangle Park, NC 27709, USA
| |
Collapse
|
141
|
Ma Q, Ma H, Xu F, Wang X, Sun W. Microfluidics in cardiovascular disease research: state of the art and future outlook. MICROSYSTEMS & NANOENGINEERING 2021; 7:19. [PMID: 34567733 PMCID: PMC8433381 DOI: 10.1038/s41378-021-00245-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/08/2021] [Accepted: 01/16/2021] [Indexed: 05/21/2023]
Abstract
Due to extremely severe morbidity and mortality worldwide, it is worth achieving a more in-depth and comprehensive understanding of cardiovascular diseases. Tremendous effort has been made to replicate the cardiovascular system and investigate the pathogenesis, diagnosis and treatment of cardiovascular diseases. Microfluidics can be used as a versatile primary strategy to achieve a holistic picture of cardiovascular disease. Here, a brief review of the application of microfluidics in comprehensive cardiovascular disease research is presented, with specific discussions of the characteristics of microfluidics for investigating cardiovascular diseases integrally, including the study of pathogenetic mechanisms, the development of accurate diagnostic methods and the establishment of therapeutic treatments. Investigations of critical pathogenetic mechanisms for typical cardiovascular diseases by microfluidic-based organ-on-a-chip are categorized and reviewed, followed by a detailed summary of microfluidic-based accurate diagnostic methods. Microfluidic-assisted cardiovascular drug evaluation and screening as well as the fabrication of novel delivery vehicles are also reviewed. Finally, the challenges with and outlook on further advancing the use of microfluidics technology in cardiovascular disease research are highlighted and discussed.
Collapse
Affiliation(s)
- Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao, 266071 China
| | - Haixia Ma
- Center for Prenatal Diagnosis, Zibo Maternal and Child Health Care Hospital, Zibo, 255000 China
| | - Fenglan Xu
- Department of Clinical Pharmacy, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001 China
| | - Xinyu Wang
- Institute of Thermal Science and Technology, Shandong University, Jinan, 250061 China
| | - Wentao Sun
- Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & School of Medicine, Nankai University, Tianjin, 300457 China
| |
Collapse
|
142
|
Yip JK, Sarkar D, Petersen AP, Gipson JN, Tao J, Kale S, Rexius-Hall ML, Cho N, Khalil NN, Kapadia R, McCain ML. Contact photolithography-free integration of patterned and semi-transparent indium tin oxide stimulation electrodes into polydimethylsiloxane-based heart-on-a-chip devices for streamlining physiological recordings. LAB ON A CHIP 2021; 21:674-687. [PMID: 33439202 PMCID: PMC7968549 DOI: 10.1039/d0lc00948b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Controlled electrical stimulation is essential for evaluating the physiology of cardiac tissues engineered in heart-on-a-chip devices. However, existing stimulation techniques, such as external platinum electrodes or opaque microelectrode arrays patterned on glass substrates, have limited throughput, reproducibility, or compatibility with other desirable features of heart-on-a-chip systems, such as the use of tunable culture substrates, imaging accessibility, or enclosure in a microfluidic device. In this study, indium tin oxide (ITO), a conductive, semi-transparent, and biocompatible material, was deposited onto glass and polydimethylsiloxane (PDMS)-coated coverslips as parallel or point stimulation electrodes using laser-cut tape masks. ITO caused substrate discoloration but did not prevent brightfield imaging. ITO-patterned substrates were microcontact printed with arrayed lines of fibronectin and seeded with neonatal rat ventricular myocytes, which assembled into aligned cardiac tissues. ITO deposited as parallel or point electrodes was connected to an external stimulator and used to successfully stimulate micropatterned cardiac tissues to generate calcium transients or propagating calcium waves, respectively. ITO electrodes were also integrated into the cantilever-based muscular thin film (MTF) assay to stimulate and quantify the contraction of micropatterned cardiac tissues. To demonstrate the potential for multiple ITO electrodes to be integrated into larger, multiplexed systems, two sets of ITO electrodes were deposited onto a single substrate and used to stimulate the contraction of distinct micropatterned cardiac tissues independently. Collectively, these approaches for integrating ITO electrodes into heart-on-a-chip devices are relatively facile, modular, and scalable and could have diverse applications in microphysiological systems of excitable tissues.
Collapse
Affiliation(s)
- Joycelyn K Yip
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Debarghya Sarkar
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Jennifer N Gipson
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Jun Tao
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Salil Kale
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Megan L Rexius-Hall
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Rehan Kapadia
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA. and Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
143
|
Jæger KH, Wall S, Tveito A. Computational prediction of drug response in short QT syndrome type 1 based on measurements of compound effect in stem cell-derived cardiomyocytes. PLoS Comput Biol 2021; 17:e1008089. [PMID: 33591962 PMCID: PMC7909705 DOI: 10.1371/journal.pcbi.1008089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/26/2021] [Accepted: 12/20/2020] [Indexed: 12/20/2022] Open
Abstract
Short QT (SQT) syndrome is a genetic cardiac disorder characterized by an abbreviated QT interval of the patient's electrocardiogram. The syndrome is associated with increased risk of arrhythmia and sudden cardiac death and can arise from a number of ion channel mutations. Cardiomyocytes derived from induced pluripotent stem cells generated from SQT patients (SQT hiPSC-CMs) provide promising platforms for testing pharmacological treatments directly in human cardiac cells exhibiting mutations specific for the syndrome. However, a difficulty is posed by the relative immaturity of hiPSC-CMs, with the possibility that drug effects observed in SQT hiPSC-CMs could be very different from the corresponding drug effect in vivo. In this paper, we apply a multistep computational procedure for translating measured drug effects from these cells to human QT response. This process first detects drug effects on individual ion channels based on measurements of SQT hiPSC-CMs and then uses these results to estimate the drug effects on ventricular action potentials and QT intervals of adult SQT patients. We find that the procedure is able to identify IC50 values in line with measured values for the four drugs quinidine, ivabradine, ajmaline and mexiletine. In addition, the predicted effect of quinidine on the adult QT interval is in good agreement with measured effects of quinidine for adult patients. Consequently, the computational procedure appears to be a useful tool for helping predicting adult drug responses from pure in vitro measurements of patient derived cell lines.
Collapse
MESH Headings
- Action Potentials/drug effects
- Adult
- Ajmaline/pharmacology
- Algorithms
- Anti-Arrhythmia Agents/pharmacology
- Arrhythmias, Cardiac/drug therapy
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Cell Line
- Computational Biology
- Drug Evaluation, Preclinical/methods
- Drug Evaluation, Preclinical/statistics & numerical data
- ERG1 Potassium Channel/genetics
- Electrocardiography
- Heart Conduction System/abnormalities
- Heart Conduction System/physiopathology
- Heart Defects, Congenital/drug therapy
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/physiopathology
- Humans
- In Vitro Techniques
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/physiology
- Ivabradine/pharmacology
- Mexiletine/pharmacology
- Models, Cardiovascular
- Mutation
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/physiology
- Quinidine/pharmacology
- Translational Research, Biomedical
Collapse
Affiliation(s)
| | | | - Aslak Tveito
- Simula Research Laboratory, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| |
Collapse
|
144
|
Liu B, Wang B, Zhang X, Lock R, Nash T, Vunjak-Novakovic G. Cell type-specific microRNA therapies for myocardial infarction. Sci Transl Med 2021; 13:eabd0914. [PMID: 33568517 PMCID: PMC8848299 DOI: 10.1126/scitranslmed.abd0914] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
Current interventions fail to recover injured myocardium after infarction and prompt the need for development of cardioprotective strategies. Of increasing interest is the therapeutic use of microRNAs to control gene expression through specific targeting of mRNAs. In this Review, we discuss current microRNA-based therapeutic strategies, describing the outcomes and limitations of key microRNAs with a focus on target cell types and molecular pathways. Last, we offer a perspective on the outlook of microRNA therapies for myocardial infarction, highlighting the outstanding challenges and emerging strategies.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bryan Wang
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor Nash
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| |
Collapse
|
145
|
Yin F, Zhang X, Wang L, Wang Y, Zhu Y, Li Z, Tao T, Chen W, Yu H, Qin J. HiPSC-derived multi-organoids-on-chip system for safety assessment of antidepressant drugs. LAB ON A CHIP 2021; 21:571-581. [PMID: 33319899 DOI: 10.1039/d0lc00921k] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The poor predictive power of existing preclinical models has spurred efforts to develop human-relevant models for accurate assessment of drug safety. In this work, we developed a multi-organoids-on-a-chip system derived from human induced pluripotent stem cells (hiPSCs), which allows for the assessment of the cardiac safety of an antidepressant drug, following liver metabolism in vitro. This liver-heart organoids-on-chip device contains compartmentalized chambers separated by a porous membrane, which permits the co-culture of 3D human liver organoids in the upper multi-well chamber and cardiac organoids in the bottom micropillar array simultaneously. The co-cultured liver and heart organoids on chip maintained good viability and human organ-specific functions respectively, including the synthesis of albumin and urea of liver organoids, and the beating function of cardiac organoids. In particular, the liver organoids displayed proper metabolic capabilities with high expression of CYP450 enzyme genes. Clomipramine, a widely used antidepressant drug, can be metabolized into an active metabolite (desmethylclomipramine) through the hepatic CYP450 enzymes of liver organoids on chip identified by mass spectrometry. After exposure to 1 μM clomipramine in the liver chamber for 24 h and 48 h, the co-cultured heart organoids in the bottom layer showed significantly reduced cell viability, impaired functions of cardiac beating and calcium flux, indicating the hepatic metabolism-dependent cardiotoxicity induced by clomipramine. By combining stem cell biology and microengineered technology, this proposed hiPSC-derived multi-organoids-on-a-chip system can reflect human organ-specific functions, as well as the complex process of drug metabolism and responses at the multi-organ level. It may provide a novel platform for the assessment of drug effectiveness and safety in vitro.
Collapse
Affiliation(s)
- Fangchao Yin
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Xu Zhang
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Li Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yaqing Wang
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Yujuan Zhu
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Zhongyu Li
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Tingting Tao
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Wenwen Chen
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Hao Yu
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Jianhua Qin
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China and Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
146
|
Visone R, Ugolini GS, Cruz-Moreira D, Marzorati S, Piazza S, Pesenti E, Redaelli A, Moretti M, Occhetta P, Rasponi M. Micro-electrode channel guide (µECG) technology: an online method for continuous electrical recording in a human beating heart-on-chip. Biofabrication 2021; 13. [PMID: 33561845 DOI: 10.1088/1758-5090/abe4c4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
Cardiac toxicity still represents a common adverse outcome causing drug attrition and post-marketing withdrawal. The development of relevant in vitro models resembling the human heart recently opened the path towards a more accurate detection of drug-induced human cardiac toxicity early in the drug development process. Organs-on-chip (OoC) have been proposed as promising tools to recapitulate in vitro the key aspects of the in vivo cardiac physiology and to provide a means to directly analyze functional readouts. In this scenario, a new device capable of continuous monitoring of electrophysiological signals from functional in vitro human hearts-on-chip is here presented. The development of cardiac microtissues was achieved through a recently published method to control the mechanical environment, while the introduction of a technology consisting in micro-electrode coaxial guides (µECG) allowed to conduct direct and non-destructive electrophysiology studies. The generated human cardiac microtissues exhibited synchronous spontaneous beating, as demonstrated by multi-point and continuous acquisition of cardiac field potential, and expression of relevant genes encoding for cardiac ion-channels. A proof-of-concept pharmacological validation on 3 drugs proved the proposed model to potentially be a powerful tool to evaluate functional cardiac toxicity.
Collapse
Affiliation(s)
- Roberta Visone
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Giovanni Stefano Ugolini
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Daniela Cruz-Moreira
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Simona Marzorati
- Translational Medicine, Accelera Srl, via Pasteur, Nerviano, Nerviano, MI, 20100, ITALY
| | - Stefano Piazza
- BiomimX Srl, Via Giovanni Durando 38/A, Milan, 20158, ITALY
| | | | - Alberto Redaelli
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Galeazzi Orthopaedic Institute, via R Galeazzi 4, Milan, 20161, ITALY
| | - Paola Occhetta
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| | - Marco Rasponi
- Politecnico di Milano Dipartimento di Elettronica Informazione e Bioingegneria, Via Ponzio 34/5, Milano, Lombardia, 20133, ITALY
| |
Collapse
|
147
|
Jæger KH, Charwat V, Wall S, Healy KE, Tveito A. Identifying Drug Response by Combining Measurements of the Membrane Potential, the Cytosolic Calcium Concentration, and the Extracellular Potential in Microphysiological Systems. Front Pharmacol 2021; 11:569489. [PMID: 33628168 PMCID: PMC7898238 DOI: 10.3389/fphar.2020.569489] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/16/2020] [Indexed: 01/01/2023] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) offer a new means to study and understand the human cardiac action potential, and can give key insight into how compounds may interact with important molecular pathways to destabilize the electrical function of the heart. Important features of the action potential can be readily measured using standard experimental techniques, such as the use of voltage sensitive dyes and fluorescent genetic reporters to estimate transmembrane potentials and cytosolic calcium concentrations. Using previously introduced computational procedures, such measurements can be used to estimate the current density of major ion channels present in hiPSC-CMs, and how compounds may alter their behavior. However, due to the limitations of optical recordings, resolving the sodium current remains difficult from these data. Here we show that if these optical measurements are complemented with observations of the extracellular potential using multi electrode arrays (MEAs), we can accurately estimate the current density of the sodium channels. This inversion of the sodium current relies on observation of the conduction velocity which turns out to be straightforwardly computed using measurements of extracellular waves across the electrodes. The combined data including the membrane potential, the cytosolic calcium concentration and the extracellular potential further opens up for the possibility of accurately estimating the effect of novel drugs applied to hiPSC-CMs.
Collapse
Affiliation(s)
| | - Verena Charwat
- Department of Bioengineering, University of California, Berkeley, CA, United States
| | | | - Kevin E. Healy
- Department of Bioengineering, University of California, Berkeley, CA, United States
- Department of Material Science and Engineering, University of California, Berkeley, CA, United States
| | - Aslak Tveito
- Simula Research Laboratory, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| |
Collapse
|
148
|
Tadevosyan K, Iglesias-García O, Mazo MM, Prósper F, Raya A. Engineering and Assessing Cardiac Tissue Complexity. Int J Mol Sci 2021; 22:ijms22031479. [PMID: 33540699 PMCID: PMC7867236 DOI: 10.3390/ijms22031479] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiac tissue engineering is very much in a current focus of regenerative medicine research as it represents a promising strategy for cardiac disease modelling, cardiotoxicity testing and cardiovascular repair. Advances in this field over the last two decades have enabled the generation of human engineered cardiac tissue constructs with progressively increased functional capabilities. However, reproducing tissue-like properties is still a pending issue, as constructs generated to date remain immature relative to native adult heart. Moreover, there is a high degree of heterogeneity in the methodologies used to assess the functionality and cardiac maturation state of engineered cardiac tissue constructs, which further complicates the comparison of constructs generated in different ways. Here, we present an overview of the general approaches developed to generate functional cardiac tissues, discussing the different cell sources, biomaterials, and types of engineering strategies utilized to date. Moreover, we discuss the main functional assays used to evaluate the cardiac maturation state of the constructs, both at the cellular and the tissue levels. We trust that researchers interested in developing engineered cardiac tissue constructs will find the information reviewed here useful. Furthermore, we believe that providing a unified framework for comparison will further the development of human engineered cardiac tissue constructs displaying the specific properties best suited for each particular application.
Collapse
Affiliation(s)
- Karine Tadevosyan
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Center for Networked Biomedical Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| |
Collapse
|
149
|
Yu J, Cai P, Chen X. Structural Regulation of Myocytes in Engineered Healthy and Diseased Cardiac Models. ACS APPLIED BIO MATERIALS 2021; 4:267-276. [DOI: 10.1021/acsabm.0c01270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jing Yu
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Pingqiang Cai
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Xiaodong Chen
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
150
|
Knowledge-Graph-Based Drug Repositioning against COVID-19 by Graph Convolutional Network with Attention Mechanism. FUTURE INTERNET 2021. [DOI: 10.3390/fi13010013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The current global crisis caused by COVID-19 almost halted normal life in most parts of the world. Due to the long development cycle for new drugs, drug repositioning becomes an effective method of screening drugs for COVID-19. To find suitable drugs for COVID-19, we add COVID-19-related information into our medical knowledge graph and utilize a knowledge-graph-based drug repositioning method to screen potential therapeutic drugs for COVID-19. Specific steps are as follows. Firstly, the information about COVID-19 is collected from the latest published literature, and gene targets of COVID-19 are added to the knowledge graph. Then, the information of COVID-19 of the knowledge graph is extracted and a drug–disease interaction prediction model based on Graph Convolutional Network with Attention (Att-GCN) is established. Att-GCN is used to extract features from the knowledge graph and the prediction matrix reconstructed through matrix operation. We evaluate the model by predicting drugs for both ordinary diseases and COVID-19. The model can achieve area under curve (AUC) of 0.954 and area under the precise recall area curve (AUPR) of 0.851 for ordinary diseases. On the drug repositioning experiment for COVID-19, five drugs predicted by the models have proved effective in clinical treatment. The experimental results confirm that the model can predict drug–disease interaction effectively for both normal diseases and COVID-19.
Collapse
|