51
|
Langhans SA. Three-Dimensional in Vitro Cell Culture Models in Drug Discovery and Drug Repositioning. Front Pharmacol 2018; 9:6. [PMID: 29410625 PMCID: PMC5787088 DOI: 10.3389/fphar.2018.00006] [Citation(s) in RCA: 971] [Impact Index Per Article: 138.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/03/2018] [Indexed: 02/06/2023] Open
Abstract
Drug development is a lengthy and costly process that proceeds through several stages from target identification to lead discovery and optimization, preclinical validation and clinical trials culminating in approval for clinical use. An important step in this process is high-throughput screening (HTS) of small compound libraries for lead identification. Currently, the majority of cell-based HTS is being carried out on cultured cells propagated in two-dimensions (2D) on plastic surfaces optimized for tissue culture. At the same time, compelling evidence suggests that cells cultured in these non-physiological conditions are not representative of cells residing in the complex microenvironment of a tissue. This discrepancy is thought to be a significant contributor to the high failure rate in drug discovery, where only a low percentage of drugs investigated ever make it through the gamut of testing and approval to the market. Thus, three-dimensional (3D) cell culture technologies that more closely resemble in vivo cell environments are now being pursued with intensity as they are expected to accommodate better precision in drug discovery. Here we will review common approaches to 3D culture, discuss the significance of 3D cultures in drug resistance and drug repositioning and address some of the challenges of applying 3D cell cultures to high-throughput drug discovery.
Collapse
Affiliation(s)
- Sigrid A. Langhans
- Nemours Center for Childhood Cancer Research and Nemours Center for Neuroscience Research, Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
52
|
Buzanska L, Zychowicz M, Kinsner-Ovaskainen A. Bioengineering of the Human Neural Stem Cell Niche: A Regulatory Environment for Cell Fate and Potential Target for Neurotoxicity. Results Probl Cell Differ 2018; 66:207-230. [PMID: 30209661 DOI: 10.1007/978-3-319-93485-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human neural stem/progenitor cells of the developing and adult organisms are surrounded by the microenvironment, so-called neurogenic niche. The developmental processes of stem cells, such as survival, proliferation, differentiation, and fate decisions, are controlled by the mutual interactions between cells and the niche components. Such interactions are tissue specific and determined by the biochemical and biophysical properties of the niche constituencies and the presence of other cell types. This dynamic approach of the stem cell niche, when translated into in vitro settings, requires building up "biomimetic" microenvironments resembling natural conditions, where the stem/progenitor cell is provided with diverse extracellular signals exerted by soluble and structural cues, mimicking those found in vivo. The neural stem cell niche is characterized by a unique composition of soluble components including neurotransmitters and trophic factors as well as insoluble extracellular matrix proteins and proteoglycans. Biotechnological innovations provide tools such as a new generation of tunable biomaterials capable of releasing specific signals in a spatially and temporally controlled manner, thus creating in vitro nature-like conditions and, when combined with stem cell-derived tissue specific progenitors, producing differentiated neuronal tissue structures. In addition, substantial progress has been made on the protocols to obtain stem cell-derived cell aggregates such as neurospheres and self-assembled organoids.In this chapter, we have assessed the application of bioengineered human neural stem cell microenvironments to produce in vitro models of different levels of biological complexity for the efficient control of stem cell fate. Examples of biomaterial-supported two-dimensional and three-dimensional (2D and 3D) complex culture systems that provide artificial neural stem cell niches are discussed in the context of their application for basic research and neurotoxicity testing.
Collapse
Affiliation(s)
- Leonora Buzanska
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland.
| | - Marzena Zychowicz
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Kinsner-Ovaskainen
- European Commission, Joint Research Centre, Directorate for Health Consumers and Reference Materials, Ispra, Italy
| |
Collapse
|
53
|
Geraili A, Jafari P, Hassani MS, Araghi BH, Mohammadi MH, Ghafari AM, Tamrin SH, Modarres HP, Kolahchi AR, Ahadian S, Sanati-Nezhad A. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms. Adv Healthc Mater 2018; 7. [PMID: 28910516 DOI: 10.1002/adhm.201700426] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/01/2017] [Indexed: 01/09/2023]
Abstract
Organ-on-chip (OOC) platforms have attracted attentions of pharmaceutical companies as powerful tools for screening of existing drugs and development of new drug candidates. OOCs have primarily used human cell lines or primary cells to develop biomimetic tissue models. However, the ability of human stem cells in unlimited self-renewal and differentiation into multiple lineages has made them attractive for OOCs. The microfluidic technology has enabled precise control of stem cell differentiation using soluble factors, biophysical cues, and electromagnetic signals. This study discusses different tissue- and organ-on-chip platforms (i.e., skin, brain, blood-brain barrier, bone marrow, heart, liver, lung, tumor, and vascular), with an emphasis on the critical role of stem cells in the synthesis of complex tissues. This study further recaps the design, fabrication, high-throughput performance, and improved functionality of stem-cell-based OOCs, technical challenges, obstacles against implementing their potential applications, and future perspectives related to different experimental platforms.
Collapse
Affiliation(s)
- Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
| | - Parya Jafari
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
- Department of Electrical Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohsen Sheikh Hassani
- Department of Systems and Computer Engineering; Carleton University; 1125 Colonel By Drive Ottawa K1S 5B6 ON Canada
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Mohammad Ghafari
- Department of Stem Cells and Developmental Biology; Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology; Tehran 16635-148 Iran
| | - Sara Hasanpour Tamrin
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
- Center for Bioengineering Research and Education; Biomedical Engineering Program; University of Calgary; Calgary T2N 1N4 AB Canada
| |
Collapse
|
54
|
Somayaji MR, Przekwas AJ, Gupta RK. Combination Therapy for Multi-Target Manipulation of Secondary Brain Injury Mechanisms. Curr Neuropharmacol 2018; 16:484-504. [PMID: 28847295 PMCID: PMC6018188 DOI: 10.2174/1570159x15666170828165711] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a major healthcare problem that affects millions of people worldwide. Despite advances in understanding and developing preventative and treatment strategies using preclinical animal models, clinical trials to date have failed, and a 'magic bullet' for effectively treating TBI-induced damage does not exist. Thus, novel pharmacological strategies to effectively manipulate the complex and heterogeneous pathophysiology of secondary injury mechanisms are needed. Given that goal, this paper discusses the relevance and advantages of combination therapies (COMTs) for 'multi-target manipulation' of the secondary injury cascade by administering multiple drugs to achieve an optimal therapeutic window of opportunity (e.g., temporally broad window) and compares these regimens to monotherapies that manipulate a single target with a single drug at a given time. Furthermore, we posit that integrated mechanistic multiscale models that combine primary injury biomechanics, secondary injury mechanobiology/neurobiology, physiology, pharmacology and mathematical programming techniques could account for vast differences in the biological space and time scales and help to accelerate drug development, to optimize pharmacological COMT protocols and to improve treatment outcomes.
Collapse
Affiliation(s)
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
55
|
Phan DTT, Bender RHF, Andrejecsk JW, Sobrino A, Hachey SJ, George SC, Hughes CCW. Blood-brain barrier-on-a-chip: Microphysiological systems that capture the complexity of the blood-central nervous system interface. Exp Biol Med (Maywood) 2017; 242:1669-1678. [PMID: 28195514 PMCID: PMC5786363 DOI: 10.1177/1535370217694100] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier is a dynamic and highly organized structure that strictly regulates the molecules allowed to cross the brain vasculature into the central nervous system. The blood-brain barrier pathology has been associated with a number of central nervous system diseases, including vascular malformations, stroke/vascular dementia, Alzheimer's disease, multiple sclerosis, and various neurological tumors including glioblastoma multiforme. There is a compelling need for representative models of this critical interface. Current research relies heavily on animal models (mostly mice) or on two-dimensional (2D) in vitro models, neither of which fully capture the complexities of the human blood-brain barrier. Physiological differences between humans and mice make translation to the clinic problematic, while monolayer cultures cannot capture the inherently three-dimensional (3D) nature of the blood-brain barrier, which includes close association of the abluminal side of the endothelium with astrocyte foot-processes and pericytes. Here we discuss the central nervous system diseases associated with blood-brain barrier pathology, recent advances in the development of novel 3D blood-brain barrier -on-a-chip systems that better mimic the physiological complexity and structure of human blood-brain barrier, and provide an outlook on how these blood-brain barrier-on-a-chip systems can be used for central nervous system disease modeling. Impact statement The field of microphysiological systems is rapidly evolving as new technologies are introduced and our understanding of organ physiology develops. In this review, we focus on Blood-Brain Barrier (BBB) models, with a particular emphasis on how they relate to neurological disorders such as Alzheimer's disease, multiple sclerosis, stroke, cancer, and vascular malformations. We emphasize the importance of capturing the three-dimensional nature of the brain and the unique architecture of the BBB - something that until recently had not been well modeled by in vitro systems. Our hope is that this review will provide a launch pad for new ideas and methodologies that can provide us with truly physiological BBB models capable of yielding new insights into the function of this critical interface.
Collapse
Affiliation(s)
- Duc TT Phan
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - R Hugh F Bender
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jillian W Andrejecsk
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Agua Sobrino
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Stephanie J Hachey
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Christopher CW Hughes
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
56
|
Watson DE, Hunziker R, Wikswo JP. Fitting tissue chips and microphysiological systems into the grand scheme of medicine, biology, pharmacology, and toxicology. Exp Biol Med (Maywood) 2017; 242:1559-1572. [PMID: 29065799 DOI: 10.1177/1535370217732765] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microphysiological systems (MPS), which include engineered organoids (EOs), single organ/tissue chips (TCs), and multiple organs interconnected to create miniature in vitro models of human physiological systems, are rapidly becoming effective tools for drug development and the mechanistic understanding of tissue physiology and pathophysiology. The second MPS thematic issue of Experimental Biology and Medicine comprises 15 articles by scientists and engineers from the National Institutes of Health, the IQ Consortium, the Food and Drug Administration, and Environmental Protection Agency, an MPS company, and academia. Topics include the progress, challenges, and future of organs-on-chips, dissemination of TCs into Pharma, children's health protection, liver zonation, liver chips and their coupling to interconnected systems, gastrointestinal MPS, maturation of immature cardiomyocytes in a heart-on-a-chip, coculture of multiple cell types in a human skin construct, use of synthetic hydrogels to create EOs that form neural tissue models, the blood-brain barrier-on-a-chip, MPS models of coupled female reproductive organs, coupling MPS devices to create a body-on-a-chip, and the use of a microformulator to recapitulate endocrine circadian rhythms. While MPS hardware has been relatively stable since the last MPS thematic issue, there have been significant advances in cell sourcing, with increased reliance on human-induced pluripotent stem cells, and in characterization of the genetic and functional cell state in MPS bioreactors. There is growing appreciation of the need to minimize perfusate-to-cell-volume ratios and respect physiological scaling of coupled TCs. Questions asked by drug developers are followed by an analysis of the potential value, costs, and needs of Pharma. Of highest value and lowest switching costs may be the development of MPS disease models to aid in the discovery of disease mechanisms; novel compounds including probes, leads, and clinical candidates; and mechanism of action of drug candidates. Impact statement Microphysiological systems (MPS), which include engineered organoids and both individual and coupled organs-on-chips and tissue chips, are a rapidly growing topic of research that addresses the known limitations of conventional cellular monoculture on flat plastic - a well-perfected set of techniques that produces reliable, statistically significant results that may not adequately represent human biology and disease. As reviewed in this article and the others in this thematic issue, MPS research has made notable progress in the past three years in both cell sourcing and characterization. As the field matures, currently identified challenges are being addressed, and new ones are being recognized. Building upon investments by the Defense Advanced Research Projects Agency, National Institutes of Health, Food and Drug Administration, Defense Threat Reduction Agency, and Environmental Protection Agency of more than $200 million since 2012 and sizable corporate spending, academic and commercial players in the MPS community are demonstrating their ability to meet the translational challenges required to apply MPS technologies to accelerate drug development and advance toxicology.
Collapse
Affiliation(s)
| | - Rosemarie Hunziker
- 2 National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Wikswo
- 3 Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics & Astronomy, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1807, USA
| |
Collapse
|
57
|
Huang G, Li F, Zhao X, Ma Y, Li Y, Lin M, Jin G, Lu TJ, Genin GM, Xu F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem Rev 2017; 117:12764-12850. [PMID: 28991456 PMCID: PMC6494624 DOI: 10.1021/acs.chemrev.7b00094] [Citation(s) in RCA: 500] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell microenvironment has emerged as a key determinant of cell behavior and function in development, physiology, and pathophysiology. The extracellular matrix (ECM) within the cell microenvironment serves not only as a structural foundation for cells but also as a source of three-dimensional (3D) biochemical and biophysical cues that trigger and regulate cell behaviors. Increasing evidence suggests that the 3D character of the microenvironment is required for development of many critical cell responses observed in vivo, fueling a surge in the development of functional and biomimetic materials for engineering the 3D cell microenvironment. Progress in the design of such materials has improved control of cell behaviors in 3D and advanced the fields of tissue regeneration, in vitro tissue models, large-scale cell differentiation, immunotherapy, and gene therapy. However, the field is still in its infancy, and discoveries about the nature of cell-microenvironment interactions continue to overturn much early progress in the field. Key challenges continue to be dissecting the roles of chemistry, structure, mechanics, and electrophysiology in the cell microenvironment, and understanding and harnessing the roles of periodicity and drift in these factors. This review encapsulates where recent advances appear to leave the ever-shifting state of the art, and it highlights areas in which substantial potential and uncertainty remain.
Collapse
Affiliation(s)
- Guoyou Huang
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Chemistry, School of Science,
Xi’an Jiaotong University, Xi’an 710049, People’s Republic
of China
| | - Xin Zhao
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Interdisciplinary Division of Biomedical
Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong,
People’s Republic of China
| | - Yufei Ma
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Yuhui Li
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Min Lin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Guorui Jin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- MOE Key Laboratory for Multifunctional Materials
and Structures, Xi’an Jiaotong University, Xi’an 710049,
People’s Republic of China
| | - Guy M. Genin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Mechanical Engineering &
Materials Science, Washington University in St. Louis, St. Louis 63130, MO,
USA
- NSF Science and Technology Center for
Engineering MechanoBiology, Washington University in St. Louis, St. Louis 63130,
MO, USA
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| |
Collapse
|
58
|
Zander NE, Piehler T, Hogberg H, Pamies D. Explosive Blast Loading on Human 3D Aggregate Minibrains. Cell Mol Neurobiol 2017; 37:1331-1334. [PMID: 28110483 PMCID: PMC11482198 DOI: 10.1007/s10571-017-0463-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/09/2017] [Indexed: 11/26/2022]
Abstract
The effects of primary explosive blast on brain tissue still remain mostly unknown. There are few in vitro models that use real explosives to probe the mechanisms of injury at the cellular level. In this work, 3D aggregates of human brain cells or brain microphysiological system were exposed to military explosives at two different pressures (50 and 100 psi). Results indicate that membrane damage and oxidative stress increased with blast pressure, but cell death remained minimal.
Collapse
Affiliation(s)
- Nicole E Zander
- Department of the Army, US Army Research Laboratory, Weapons and Materials Research Directorate, RDRL-WMM-G, Building 4600, Aberdeen Proving Ground, MD, 21005, USA.
| | - Thuvan Piehler
- Department of the Army, US Army Research Laboratory, Weapons and Materials Research Directorate, RDRL-WMM-G, Building 4600, Aberdeen Proving Ground, MD, 21005, USA
| | - Helena Hogberg
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615N. Wolfe Street, Baltimore, MD, 21205, USA
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615N. Wolfe Street, Baltimore, MD, 21205, USA
| |
Collapse
|
59
|
Choi JH, Cho HY, Choi JW. Microdevice Platform for In Vitro Nervous System and Its Disease Model. Bioengineering (Basel) 2017; 4:E77. [PMID: 28952555 PMCID: PMC5615323 DOI: 10.3390/bioengineering4030077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 01/09/2023] Open
Abstract
The development of precise microdevices can be applied to the reconstruction of in vitro human microenvironmental systems with biomimetic physiological conditions that have highly tunable spatial and temporal features. Organ-on-a-chip can emulate human physiological functions, particularly at the organ level, as well as its specific roles in the body. Due to the complexity of the structure of the central nervous system and its intercellular interaction, there remains an urgent need for the development of human brain or nervous system models. Thus, various microdevice models have been proposed to mimic actual human brain physiology, which can be categorized as nervous system-on-a-chip. Nervous system-on-a-chip platforms can prove to be promising technologies, through the application of their biomimetic features to the etiology of neurodegenerative diseases. This article reviews the microdevices for nervous system-on-a-chip platform incorporated with neurobiology and microtechnology, including microfluidic designs that are biomimetic to the entire nervous system. The emulation of both neurodegenerative disorders and neural stem cell behavior patterns in micro-platforms is also provided, which can be used as a basis to construct nervous system-on-a-chip.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea.
| | - Hyeon-Yeol Cho
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea.
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 610 Taylor Road, Piscataway, NJ 08854, USA.
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea.
| |
Collapse
|
60
|
Abstract
Traumatic Brain Injury (TBI) remains a significant cause of mortality and morbidity, affecting individuals of all age groups. Much remains to be learned about its complex pathophysiology, with a view to designing effective neuroprotective strategies to protect sublethally injured brain tissue that would otherwise die in secondary injury processes. Experimental in vivo models offer the potential to study TBI in the laboratory, however, treatments that were neuroprotective in animals have, thus far, largely failed to translate in human clinical studies. In vitro models of neurotrauma can be used to study specific pathophysiological cascades — individually and without confounding factors — and to test potential neuroprotective strategies. These in vitro models include transection, compression, barotrauma, acceleration, hydrodynamic, chemical injury and cell-stretch methodologies. Various cell culture systems can also be utilised, including brain-on-a-chip, immortalised cell lines, primary cultures, acute preparations and organotypic cultures. Potential positive outcomes of the increased use of in vitro platforms to study TBI would be the refinement of in vivo experiments, as well as enhanced translation of the results into clinically meaningful neuroprotective strategies for the future. In addition, the replacement of in vivo experiments by suitable in vitro studies would lead to a welcome reduction in the numbers of animal procedures in this ethically-challenging field.
Collapse
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Queen's Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
61
|
Knudsen TB, Klieforth B, Slikker W. Programming microphysiological systems for children's health protection. Exp Biol Med (Maywood) 2017; 242:1586-1592. [PMID: 28658972 DOI: 10.1177/1535370217717697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Microphysiological systems (MPS) and computer simulation models that recapitulate the underlying biology and toxicology of critical developmental transitions are emerging tools for developmental effects assessment of drugs/chemicals. Opportunities and challenges exist for their application to alternative, more public health relevant and efficient chemical toxicity testing methods. This is especially pertinent to children's health research and the evaluation of complex embryological and reproductive impacts of drug/chemical exposure. Scaling these technologies to higher throughput is a key challenge and drives the need for in silico models for quantitative prediction of developmental toxicity to inform safety assessments. One example is cellular agent-based models, constructed from extant embryology, that produce data useful to simulate critical developmental transitions and thereby predict phenotypic consequences of disruption in silico. Biologically inspired MPS models built from human induced pluripotent stem (iPS)-derived cells and synthetic matrices that recapitulate organ-specific physiologies and native tissue architectures are providing exciting new research opportunities to advance the assessment of developmental toxicity and offer the possibility of deriving a full 'human on a chip' system, or a 'Homunculus.' Impact statement This 'commentary' summarizes research needs and opportunities for engineered MPS models for developmental and reproductive toxicity testing. Emerging concepts can be taken forward to a virtual tissue modeling framework for assessing chemical (and non-chemical) stressors on human development. These models will advance children's health research, both basic and translational and new ways to evaluate complex embryological and reproductive impacts of drug and chemical exposures to inform safety assessments.
Collapse
Affiliation(s)
- T B Knudsen
- 1 National Center for Computational Toxicology/EPA, Research Triangle Park, NC 27711, USA
| | - B Klieforth
- 2 National Center for Environmental Research/EPA, Washington, DC 20460, USA
| | - W Slikker
- 3 National Center for Toxicological Research/FDA, Jefferson, AR 72079, USA
| |
Collapse
|
62
|
Haring AP, Sontheimer H, Johnson BN. Microphysiological Human Brain and Neural Systems-on-a-Chip: Potential Alternatives to Small Animal Models and Emerging Platforms for Drug Discovery and Personalized Medicine. Stem Cell Rev Rep 2017; 13:381-406. [PMID: 28488234 PMCID: PMC5534264 DOI: 10.1007/s12015-017-9738-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Translational challenges associated with reductionist modeling approaches, as well as ethical concerns and economic implications of small animal testing, drive the need for developing microphysiological neural systems for modeling human neurological diseases, disorders, and injuries. Here, we provide a comprehensive review of microphysiological brain and neural systems-on-a-chip (NSCs) for modeling higher order trajectories in the human nervous system. Societal, economic, and national security impacts of neurological diseases, disorders, and injuries are highlighted to identify critical NSC application spaces. Hierarchical design and manufacturing of NSCs are discussed with distinction for surface- and bulk-based systems. Three broad NSC classes are identified and reviewed: microfluidic NSCs, compartmentalized NSCs, and hydrogel NSCs. Emerging areas and future directions are highlighted, including the application of 3D printing to design and manufacturing of next-generation NSCs, the use of stem cells for constructing patient-specific NSCs, and the application of human NSCs to 'personalized neurology'. Technical hurdles and remaining challenges are discussed. This review identifies the state-of-the-art design methodologies, manufacturing approaches, and performance capabilities of NSCs. This work suggests NSCs appear poised to revolutionize the modeling of human neurological diseases, disorders, and injuries.
Collapse
Affiliation(s)
- Alexander P Haring
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Virginia Tech Carilion Research Institute, Roanoke, VA, 24016, USA
- School of Neuroscience, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Blake N Johnson
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
- School of Neuroscience, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Materials Science and Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
63
|
Malinovskaya NA, Komleva YK, Salmin VV, Morgun AV, Shuvaev AN, Panina YA, Boitsova EB, Salmina AB. Endothelial Progenitor Cells Physiology and Metabolic Plasticity in Brain Angiogenesis and Blood-Brain Barrier Modeling. Front Physiol 2016; 7:599. [PMID: 27990124 PMCID: PMC5130982 DOI: 10.3389/fphys.2016.00599] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/16/2016] [Indexed: 12/31/2022] Open
Abstract
Currently, there is a considerable interest to the assessment of blood-brain barrier (BBB) development as a part of cerebral angiogenesis developmental program. Embryonic and adult angiogenesis in the brain is governed by the coordinated activity of endothelial progenitor cells, brain microvascular endothelial cells, and non-endothelial cells contributing to the establishment of the BBB (pericytes, astrocytes, neurons). Metabolic and functional plasticity of endothelial progenitor cells controls their timely recruitment, precise homing to the brain microvessels, and efficient support of brain angiogenesis. Deciphering endothelial progenitor cells physiology would provide novel engineering approaches to establish adequate microfluidically-supported BBB models and brain microphysiological systems for translational studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alla B. Salmina
- Research Institute of Molecular Medicine & Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| |
Collapse
|
64
|
Vagaska B, Ferretti P. Toward modeling the human nervous system in a dish: recent progress and outstanding challenges. Regen Med 2016; 12:15-23. [PMID: 27900887 DOI: 10.2217/rme-2016-0106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Studying the cellular and molecular bases governing development, and normal and abnormal functions of the human CNS is hampered by its complexity and the very limited possibility of experimentally manipulating it in vivo. Development of 3D, tissue-like culture systems offers much promise for boosting our understanding of human neural development, birth defects, neurodegenerative diseases and neural injury, and for providing platforms that will more accurately predict efficacy of putative therapeutic compounds and assess responses to potentially neurotoxic agents. Although novel technological developments and a more interdisciplinary approach to modeling the human CNS are accelerating the pace of discovery, increasing the complexity of in vitro systems increases the ordeals to be overcome to establish highly reproducible models amenable to quantitative analysis.
Collapse
Affiliation(s)
- Barbora Vagaska
- Stem Cell & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Patrizia Ferretti
- Stem Cell & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| |
Collapse
|
65
|
Towards a 21st-century roadmap for biomedical research and drug discovery: consensus report and recommendations. Drug Discov Today 2016; 22:327-339. [PMID: 27989722 DOI: 10.1016/j.drudis.2016.10.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/15/2016] [Accepted: 10/24/2016] [Indexed: 02/08/2023]
Abstract
Decades of costly failures in translating drug candidates from preclinical disease models to human therapeutic use warrant reconsideration of the priority placed on animal models in biomedical research. Following an international workshop attended by experts from academia, government institutions, research funding bodies, and the corporate and non-governmental organisation (NGO) sectors, in this consensus report, we analyse, as case studies, five disease areas with major unmet needs for new treatments. In view of the scientifically driven transition towards a human pathways-based paradigm in toxicology, a similar paradigm shift appears to be justified in biomedical research. There is a pressing need for an approach that strategically implements advanced, human biology-based models and tools to understand disease pathways at multiple biological scales. We present recommendations to help achieve this.
Collapse
|
66
|
Development and characterization of a human embryonic stem cell-derived 3D neural tissue model for neurotoxicity testing. Toxicol In Vitro 2016; 38:124-135. [PMID: 27729293 DOI: 10.1016/j.tiv.2016.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 02/08/2023]
Abstract
Alternative models for more rapid compound safety testing are of increasing demand. With emerging techniques using human pluripotent stem cells, the possibility of generating human in vitro models has gained interest, as factors related to species differences could be potentially eliminated. When studying potential neurotoxic effects of a compound it is of crucial importance to have both neurons and glial cells. We have successfully developed a protocol for generating in vitro 3D human neural tissues, using neural progenitor cells derived from human embryonic stem cells. These 3D neural tissues can be maintained for two months and undergo progressive differentiation. We showed a gradual decreased expression of early neural lineage markers, paralleled by an increase in markers specific for mature neurons, astrocytes and oligodendrocytes. At the end of the two-month culture period the neural tissues not only displayed synapses and immature myelin sheaths around axons, but electrophysiological measurements also showed spontaneous activity. Neurotoxicity testing - comparing non-neurotoxic to known neurotoxic model compounds - showed an expected increase in the marker of astroglial reactivity after exposure to known neurotoxicants methylmercury and trimethyltin. Although further characterization and refinement of the model is required, these results indicate its potential usefulness for in vitro neurotoxicity testing.
Collapse
|
67
|
Lee H, Kim DS, Ha SK, Choi I, Lee JM, Sung JH. A pumpless multi-organ-on-a-chip (MOC) combined with a pharmacokinetic-pharmacodynamic (PK-PD) model. Biotechnol Bioeng 2016; 114:432-443. [PMID: 27570096 DOI: 10.1002/bit.26087] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/09/2016] [Accepted: 08/21/2016] [Indexed: 12/15/2022]
Abstract
A multi-organ-on-a-chip (MOC), also known as a human-on-a-chip, aims to simulate whole body response to drugs by connecting microscale cell cultures of multiple tissue types via fluidic channels and reproducing the interaction between them. While several studies have demonstrated the usefulness of MOC at a proof-of-concept level, improvements are needed to enable wider acceptance of such systems; ease of use for general biological researchers, and a mathematical framework to design and interpret the MOC systems. Here, we introduce a pumpless, user-friendly MOC which can be easily assembled and operated, and demonstrate the use of a PK-PD model for interpreting drug's action inside the MOC. The metabolism-dependent anticancer activity of a flavonoid, luteolin, was evaluated in a two-compartment MOC containing the liver (HepG2) and the tumor (HeLa) cells, and the observed anticancer activity was significantly weaker than that anticipated from a well plate study. Simulation of a PK-PD model revealed that simultaneous metabolism and tumor-killing actions likely resulted in a decreased anti-cancer effect. Our work demonstrates that the combined platform of mathematical PK-PD model and an experimental MOC can be a useful tool for gaining an insight into the mechanism of action of drugs with interactions between multiple organs. Biotechnol. Bioeng. 2017;114: 432-443. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hyuna Lee
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| | - Dae Shik Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Sang Keun Ha
- Korea Food Research Institute, Seongnam-si, Gyenggi-do, Republic of Korea
| | - Inwook Choi
- Korea Food Research Institute, Seongnam-si, Gyenggi-do, Republic of Korea
| | - Jong Min Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| |
Collapse
|
68
|
Abstract
The exponential growth of the Internet of Things and the global popularity and remarkable decline in cost of the mobile phone is driving the digital transformation of medical practice. The rapidly maturing digital, non-medical world of mobile (wireless) devices, cloud computing and social networking is coalescing with the emerging digital medical world of omics data, biosensors and advanced imaging which offers the increasingly realistic prospect of personalized medicine. Described as a potential “seismic” shift from the current “healthcare” model to a “wellness” paradigm that is predictive, preventative, personalized and participatory, this change is based on the development of increasingly sophisticated biosensors which can track and measure key biochemical variables in people. Additional key drivers in this shift are metabolomic and proteomic signatures, which are increasingly being reported as pre-symptomatic, diagnostic and prognostic of toxicity and disease. These advancements also have profound implications for toxicological evaluation and safety assessment of pharmaceuticals and environmental chemicals. An approach based primarily on human in vivo and high-throughput in vitro human cell-line data is a distinct possibility. This would transform current chemical safety assessment practice which operates in a human “data poor” to a human “data rich” environment. This could also lead to a seismic shift from the current animal-based to an animal-free chemical safety assessment paradigm.
Collapse
Affiliation(s)
- George D Loizou
- Health Risks, Health and Safety Laboratory, Health and Safety Executive Buxton, UK
| |
Collapse
|
69
|
Gori M, Simonelli MC, Giannitelli SM, Businaro L, Trombetta M, Rainer A. Investigating Nonalcoholic Fatty Liver Disease in a Liver-on-a-Chip Microfluidic Device. PLoS One 2016; 11:e0159729. [PMID: 27438262 PMCID: PMC4954713 DOI: 10.1371/journal.pone.0159729] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/07/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIM Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease worldwide, ranging from simple steatosis to nonalcoholic steatohepatitis, which may progress to cirrhosis, eventually leading to hepatocellular carcinoma (HCC). HCC ranks as the third highest cause of cancer-related death globally, requiring an early diagnosis of NAFLD as a potential risk factor. However, the molecular mechanisms underlying NAFLD are still under investigation. So far, many in vitro studies on NAFLD have been hampered by the limitations of 2D culture systems, in which cells rapidly lose tissue-specific functions. The present liver-on-a-chip approach aims at filling the gap between conventional in vitro models, often scarcely predictive of in vivo conditions, and animal models, potentially biased by their xenogeneic nature. METHODS HepG2 cells were cultured into a microfluidically perfused device under free fatty acid (FFA) supplementation, namely palmitic and oleic acid, for 24h and 48h. The device mimicked the endothelial-parenchymal interface of a liver sinusoid, allowing the diffusion of nutrients and removal of waste products similar to the hepatic microvasculature. Assessment of intracellular lipid accumulation, cell viability/cytotoxicity and oxidative stress due to the FFA overload, was performed by high-content analysis methodologies using fluorescence-based functional probes. RESULTS The chip enables gradual and lower intracellular lipid accumulation, higher hepatic cell viability and minimal oxidative stress in microfluidic dynamic vs. 2D static cultures, thus mimicking the chronic condition of steatosis observed in vivo more closely. CONCLUSIONS Overall, the liver-on-a-chip system provides a suitable culture microenvironment, representing a more reliable model compared to 2D cultures for investigating NAFLD pathogenesis. Hence, our system is amongst the first in vitro models of human NAFLD developed within a microfluidic device in a sinusoid-like fashion, endowing a more permissive tissue-like microenvironment for long-term culture of hepatic cells than conventional 2D static cultures.
Collapse
Affiliation(s)
- Manuele Gori
- Department of Engineering, Tissue Engineering Laboratory, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Maria Chiara Simonelli
- Department of Engineering, Tissue Engineering Laboratory, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Sara Maria Giannitelli
- Department of Engineering, Tissue Engineering Laboratory, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Luca Businaro
- National Research Council - Institute for Photonics and Nanotechnologies (CNR-IFN), Rome, Italy
- UCBM-IFN Joint Laboratory for Nanotechnologies for the Life Sciences, Rome, Italy
| | - Marcella Trombetta
- Department of Engineering, Tissue Engineering Laboratory, Università Campus Bio-Medico di Roma, Rome, Italy
- UCBM-IFN Joint Laboratory for Nanotechnologies for the Life Sciences, Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Tissue Engineering Laboratory, Università Campus Bio-Medico di Roma, Rome, Italy
- National Research Council - Institute for Photonics and Nanotechnologies (CNR-IFN), Rome, Italy
- UCBM-IFN Joint Laboratory for Nanotechnologies for the Life Sciences, Rome, Italy
- * E-mail:
| |
Collapse
|
70
|
Karimi M, Bahrami S, Mirshekari H, Basri SMM, Nik AB, Aref AR, Akbari M, Hamblin MR. Microfluidic systems for stem cell-based neural tissue engineering. LAB ON A CHIP 2016; 16:2551-71. [PMID: 27296463 PMCID: PMC4935609 DOI: 10.1039/c6lc00489j] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Neural tissue engineering aims at developing novel approaches for the treatment of diseases of the nervous system, by providing a permissive environment for the growth and differentiation of neural cells. Three-dimensional (3D) cell culture systems provide a closer biomimetic environment, and promote better cell differentiation and improved cell function, than could be achieved by conventional two-dimensional (2D) culture systems. With the recent advances in the discovery and introduction of different types of stem cells for tissue engineering, microfluidic platforms have provided an improved microenvironment for the 3D-culture of stem cells. Microfluidic systems can provide more precise control over the spatiotemporal distribution of chemical and physical cues at the cellular level compared to traditional systems. Various microsystems have been designed and fabricated for the purpose of neural tissue engineering. Enhanced neural migration and differentiation, and monitoring of these processes, as well as understanding the behavior of stem cells and their microenvironment have been obtained through application of different microfluidic-based stem cell culture and tissue engineering techniques. As the technology advances it may be possible to construct a "brain-on-a-chip". In this review, we describe the basics of stem cells and tissue engineering as well as microfluidics-based tissue engineering approaches. We review recent testing of various microfluidic approaches for stem cell-based neural tissue engineering.
Collapse
Affiliation(s)
- Mahdi Karimi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sajad Bahrami
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran. and Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hamed Mirshekari
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran.
| | - Seyed Masoud Moosavi Basri
- Bioenvironmental Research Center, Sharif University of Technology, Tehran, Iran. and Civil & Environmental Engineering Department, Shahid Beheshti University, Tehran, Iran
| | - Amirala Bakhshian Nik
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, University of Tehran, Iran.
| | - Amir R Aref
- Department of Cancer Biology, Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA and Department of Genetics, Harvard Medical School, Boston, MA 02215, USA.
| | - Mohsen Akbari
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA. and Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. and Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| |
Collapse
|
71
|
Pistollato F, Ohayon EL, Lam A, Langley GR, Novak TJ, Pamies D, Perry G, Trushina E, Williams RS, Roher AE, Hartung T, Harnad S, Barnard N, Morris MC, Lai MC, Merkley R, Chandrasekera PC. Alzheimer disease research in the 21st century: past and current failures, new perspectives and funding priorities. Oncotarget 2016; 7:38999-39016. [PMID: 27229915 PMCID: PMC5129909 DOI: 10.18632/oncotarget.9175] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
Much of Alzheimer disease (AD) research has been traditionally based on the use of animals, which have been extensively applied in an effort to both improve our understanding of the pathophysiological mechanisms of the disease and to test novel therapeutic approaches. However, decades of such research have not effectively translated into substantial therapeutic success for human patients. Here we critically discuss these issues in order to determine how existing human-based methods can be applied to study AD pathology and develop novel therapeutics. These methods, which include patient-derived cells, computational analysis and models, together with large-scale epidemiological studies represent novel and exciting tools to enhance and forward AD research. In particular, these methods are helping advance AD research by contributing multifactorial and multidimensional perspectives, especially considering the crucial role played by lifestyle risk factors in the determination of AD risk. In addition to research techniques, we also consider related pitfalls and flaws in the current research funding system. Conversely, we identify encouraging new trends in research and government policy. In light of these new research directions, we provide recommendations regarding prioritization of research funding. The goal of this document is to stimulate scientific and public discussion on the need to explore new avenues in AD research, considering outcome and ethics as core principles to reliably judge traditional research efforts and eventually undertake new research strategies.
Collapse
Affiliation(s)
| | - Elan L. Ohayon
- Green Neuroscience Laboratory, Neurolinx Research Institute, San Diego, CA, USA
| | - Ann Lam
- Physicians Committee for Responsible Medicine, Washington, DC, USA
- Green Neuroscience Laboratory, Neurolinx Research Institute, San Diego, CA, USA
| | - Gillian R. Langley
- Research and Toxicology Department, Humane Society International, London, UK
| | | | - David Pamies
- CAAT, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | | | - Robin S.B. Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Alex E. Roher
- Division of Clinical Education, Midwestern University, Glendale, AZ, USA
- Division of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Thomas Hartung
- CAAT, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stevan Harnad
- Department of Psychology, University of Quebec/Montreal, Montreal, Canada
| | - Neal Barnard
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | - Martha Clare Morris
- Section of Nutrition and Nutritional Epidemiology, Department of Internal Medicine, Rush University, Chicago, IL, USA
| | - Mei-Chun Lai
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | - Ryan Merkley
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | | |
Collapse
|
72
|
Heussner A, Paget T. Evaluation of renal in vitro models used in ochratoxin research. WORLD MYCOTOXIN J 2016. [DOI: 10.3920/wmj2015.1975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ochratoxin A (OTA) induces renal carcinomas in rodents with a specific localisation in the S3 segment of proximal tubules and distinct early severe tissue alterations, which have been observed also in other species. Pronounced species- and sex-specific differences in toxicity occur and similar effects cannot be excluded in humans, however precise mechanism(s) remain elusive until today. In such cases, the use of in vitro models for mechanistic investigations can be very useful; in particular if a non-genotoxic mechanism of cancer formation is assumed which include cytotoxic effects. However, potential genotoxic mechanisms can also be investigated in vitro. A crucial issue of in vitro research is the choice of the appropriate cell model. Apparently, the cellular target of OTA is the renal proximal tubular cell; therefore cells from this tissue area are the most reasonable model. Furthermore, cells from affected species should be used and can be compared to cells of human origin. Another important parameter is whether to use primary cultures or to choose a cell line from the huge variety of cell lines available. In any case, important characteristics and quality controls need to be verified beforehand. Therefore, this review discusses the renal in vitro models that have been used for the investigation of renal ochratoxin toxicity. In particular, we discuss the choice of the models and the essential parameters making them suitable models for ochratoxin research together with exemplary results from this research. Furthermore, new promising models such as hTERT-immortalised cells and 3D-cultures are briefly discussed.
Collapse
Affiliation(s)
- A.H. Heussner
- Human and Environmental Toxicology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
- Pharmacy Health and Well-being, University of Sunderland, Sciences Complex, Wharncliffe Street, Sunderland SR1 3SD, United Kingdom
| | - T. Paget
- Pharmacy Health and Well-being, University of Sunderland, Sciences Complex, Wharncliffe Street, Sunderland SR1 3SD, United Kingdom
| |
Collapse
|
73
|
Benfenati E, Berggren E, Fritsche E, Hartung T, Slikker W, Spielmann H, Testai E, Tice RR, Tiramani M, Villenave R. Novel chemical hazard characterisation approaches. EFSA J 2016. [DOI: 10.2903/j.efsa.2016.s0506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
| | | | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental Medicine Germany
| | | | | | | | | | - Raymond R. Tice
- National Institute of Environmental Health Sciences (NIEHS) USA
| | | | | |
Collapse
|
74
|
Sugimura R. Bioengineering Hematopoietic Stem Cell Niche toward Regenerative Medicine. Adv Drug Deliv Rev 2016; 99:212-220. [PMID: 26527127 DOI: 10.1016/j.addr.2015.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/20/2015] [Accepted: 10/15/2015] [Indexed: 12/20/2022]
Abstract
The scope of this chapter is to introduce the current consensus of hematopoietic stem cell (HSC) niche biology to bioengineering field so that can apply to regenerative medicine. A decade of research has been addressing "what is HSC niche", then next step is "how it advances medicine". The demand to improve HSC transplantation has advanced the methodology to expand HSC in vitro. Still precise modeling of bone marrow (BM) is demanded by bioengineering HSC niche in vitro. Better understanding of HSC niche is essential toward this progress. Now it would be the time to apply the knowledge of HSC niche field to the venue of bioengineering, so that a promising new approach to regenerative medicine might appear. This chapter describes the current consensus of niche that endothelial cell and perivascular mesenchymal stromal cell maintain HSC, expansion of cord blood HSC by small molecules, bioengineering efforts to model HSC niche by microfluidics chip, organoids, and breakthroughs to induce HSC from heterologous types of cells.
Collapse
|
75
|
|
76
|
van der Helm MW, van der Meer AD, Eijkel JCT, van den Berg A, Segerink LI. Microfluidic organ-on-chip technology for blood-brain barrier research. Tissue Barriers 2016; 4:e1142493. [PMID: 27141422 PMCID: PMC4836466 DOI: 10.1080/21688370.2016.1142493] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 12/28/2015] [Accepted: 01/02/2016] [Indexed: 12/25/2022] Open
Abstract
Organs-on-chips are a new class of microengineered laboratory models that combine several of the advantages of current in vivo and in vitro models. In this review, we summarize the advances that have been made in the development of organ-on-chip models of the blood-brain barrier (BBBs-on-chips) and the challenges that are still ahead. The BBB is formed by specialized e3ndothelial cells and separates blood from brain tissue. It protects the brain from harmful compounds from the blood and provides homeostasis for optimal neuronal function. Studying BBB function and dysfunction is important for drug development and biomedical research. Microfluidic BBBs-on-chips enable real-time study of (human) cells in an engineered physiological microenvironment, for example incorporating small geometries and fluid flow as well as sensors. Examples of BBBs-on-chips in literature already show the potential of more realistic microenvironments and the study of organ-level functions. A key challenge in the field of BBB-on-chip development is the current lack of standardized quantification of parameters such as barrier permeability and shear stress. This limits the potential for direct comparison of the performance of different BBB-on-chip models to each other and existing models. We give recommendations for further standardization in model characterization and conclude that the rapidly emerging field of BBB-on-chip models holds great promise for further studies in BBB biology and drug development.
Collapse
Affiliation(s)
- Marinke W van der Helm
- BIOS Lab on a Chip group, MIRA Institute for Biomedical Technology and Technical Medicine & MESA+ Institute for Nanotechnology, University of Twente ; Enschede, The Netherlands
| | - Andries D van der Meer
- Applied Stem Cell Technologies, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente ; Enschede, The Netherlands
| | - Jan C T Eijkel
- BIOS Lab on a Chip group, MIRA Institute for Biomedical Technology and Technical Medicine & MESA+ Institute for Nanotechnology, University of Twente ; Enschede, The Netherlands
| | - Albert van den Berg
- BIOS Lab on a Chip group, MIRA Institute for Biomedical Technology and Technical Medicine & MESA+ Institute for Nanotechnology, University of Twente ; Enschede, The Netherlands
| | - Loes I Segerink
- BIOS Lab on a Chip group, MIRA Institute for Biomedical Technology and Technical Medicine & MESA+ Institute for Nanotechnology, University of Twente ; Enschede, The Netherlands
| |
Collapse
|
77
|
A Dishful of a Troubled Mind: Induced Pluripotent Stem Cells in Psychiatric Research. Stem Cells Int 2015; 2016:7909176. [PMID: 26839567 PMCID: PMC4709917 DOI: 10.1155/2016/7909176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Neuronal differentiation of induced pluripotent stem cells and direct reprogramming represent powerful methods for modeling the development of neurons in vitro. Moreover, this approach is also a means for comparing various cellular phenotypes between cell lines originating from healthy and diseased individuals or isogenic cell lines engineered to differ at only one or a few genomic loci. Despite methodological constraints and initial skepticism regarding this approach, the field is expanding at a fast pace. The improvements include the development of new differentiation protocols resulting in selected neuronal populations (e.g., dopaminergic, GABAergic, hippocampal, and cortical), the widespread use of genome editing methods, and single-cell techniques. A major challenge awaiting in vitro disease modeling is the integration of clinical data in the models, by selection of well characterized clinical populations. Ideally, these models will also demonstrate how different diagnostic categories share overlapping molecular disease mechanisms, but also have unique characteristics. In this review we evaluate studies with regard to the described developments, to demonstrate how differentiation of induced pluripotent stem cells and direct reprogramming can contribute to psychiatry.
Collapse
|
78
|
Singh S, Srivastava A, Kumar V, Pandey A, Kumar D, Rajpurohit CS, Khanna VK, Yadav S, Pant AB. Stem Cells in Neurotoxicology/Developmental Neurotoxicology: Current Scenario and Future Prospects. Mol Neurobiol 2015; 53:6938-6949. [DOI: 10.1007/s12035-015-9615-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/03/2015] [Indexed: 12/26/2022]
|
79
|
Bal-Price A, Crofton KM, Leist M, Allen S, Arand M, Buetler T, Delrue N, FitzGerald RE, Hartung T, Heinonen T, Hogberg H, Bennekou SH, Lichtensteiger W, Oggier D, Paparella M, Axelstad M, Piersma A, Rached E, Schilter B, Schmuck G, Stoppini L, Tongiorgi E, Tiramani M, Monnet-Tschudi F, Wilks MF, Ylikomi T, Fritsche E. International STakeholder NETwork (ISTNET): creating a developmental neurotoxicity (DNT) testing road map for regulatory purposes. Arch Toxicol 2015; 89:269-87. [PMID: 25618548 PMCID: PMC4309915 DOI: 10.1007/s00204-015-1464-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/04/2014] [Indexed: 01/03/2023]
Abstract
A major problem in developmental neurotoxicity (DNT) risk assessment is the lack of toxicological hazard information for most compounds. Therefore, new approaches are being considered to provide adequate experimental data that allow regulatory decisions. This process requires a matching of regulatory needs on the one hand and the opportunities provided by new test systems and methods on the other hand. Alignment of academically and industrially driven assay development with regulatory needs in the field of DNT is a core mission of the International STakeholder NETwork (ISTNET) in DNT testing. The first meeting of ISTNET was held in Zurich on 23-24 January 2014 in order to explore the concept of adverse outcome pathway (AOP) to practical DNT testing. AOPs were considered promising tools to promote test systems development according to regulatory needs. Moreover, the AOP concept was identified as an important guiding principle to assemble predictive integrated testing strategies (ITSs) for DNT. The recommendations on a road map towards AOP-based DNT testing is considered a stepwise approach, operating initially with incomplete AOPs for compound grouping, and focussing on key events of neurodevelopment. Next steps to be considered in follow-up activities are the use of case studies to further apply the AOP concept in regulatory DNT testing, making use of AOP intersections (common key events) for economic development of screening assays, and addressing the transition from qualitative descriptions to quantitative network modelling.
Collapse
Affiliation(s)
- Anna Bal-Price
- Systems Toxicology Unit, EURL-ECVAM, Institute for Health and Consumer Protection, European Commission, Joint Research Centre, TP 580, Via Fermi 1, 21026, Ispra, VA, Italy,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
The new cover of Experimental Biology and Medicine features the hermeneutic circle of biology, a concept we have adapted from the hermeneutic principle that one understands the whole only in terms of each part and the parts only in terms of the whole. Our hermeneutic circle summarizes the course of experimental biology through 2500 years of the achievements of reductionist research (understanding the parts), which culminates in our ability to rapidly sequence the genome. Rather than returning along the same path in a constructionist approach that simply builds upon this knowledge, but in reverse, an alternative is to close the circle with synthetic constructions that seek to integrate the full complexity of biological and physiological systems (understanding the whole), of which organs-on-chips are one example. This closing of the circle cannot be a comprehensively accurate representation of biology, but it can be a synthetic one that effectively defines particular biological subsystems. The illustration of the hermeneutic circle of biology is also intended to suggest both the multiple cycles that may be required to reach such a synthesis and the expansion of the circle in an outward spiral as knowledge increases. Our commentary explains the symbolism of the new cover in a philosophical and scientific discussion.
Collapse
Affiliation(s)
- John P Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education, Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics & Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| | - Andrew P Porter
- Center for Theology and the Natural Sciences, Graduate Theological Union, Berkeley, CA 94709-1212, USA
| |
Collapse
|
81
|
Lairmore MD, Ilkiw J. Animals Used in Research and Education, 1966-2016: Evolving Attitudes, Policies, and Relationships. JOURNAL OF VETERINARY MEDICAL EDUCATION 2015; 42:425-440. [PMID: 26673210 DOI: 10.3138/jvme.0615-087r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Since the inception of the Association of American Veterinary Medical Colleges (AAVMC), the use of animals in research and education has been a central element of the programs of member institutions. As veterinary education and research programs have evolved over the past 50 years, so too have societal views and regulatory policies. AAVMC member institutions have continually responded to these events by exchanging best practices in training their students in the framework of comparative medicine and the needs of society. Animals provide students and faculty with the tools to learn the fundamental knowledge and skills of veterinary medicine and scientific discovery. The study of animal models has contributed extensively to medicine, veterinary medicine, and basic sciences as these disciplines seek to understand life processes. Changing societal views over the past 50 years have provided active examination and continued refinement of the use of animals in veterinary medical education and research. The future use of animals to educate and train veterinarians will likely continue to evolve as technological advances are applied to experimental design and educational systems. Natural animal models of both human and animal health will undoubtedly continue to serve a significant role in the education of veterinarians and in the development of new treatments of animal and human disease. As it looks to the future, the AAVMC as an organization will need to continue to support and promote best practices in the humane care and appropriate use of animals in both education and research.
Collapse
MESH Headings
- Animal Experimentation/history
- Animal Experimentation/legislation & jurisprudence
- Animal Use Alternatives/history
- Animal Use Alternatives/legislation & jurisprudence
- Animal Use Alternatives/trends
- Animal Welfare/history
- Animal Welfare/legislation & jurisprudence
- Animals
- Animals, Laboratory
- Education, Veterinary/history
- Education, Veterinary/methods
- Education, Veterinary/trends
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- History, Ancient
- Human-Animal Bond
- Humans
- Models, Animal
- United States
Collapse
|
82
|
Wikswo JP. The relevance and potential roles of microphysiological systems in biology and medicine. Exp Biol Med (Maywood) 2014; 239:1061-72. [PMID: 25187571 PMCID: PMC4330974 DOI: 10.1177/1535370214542068] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Microphysiological systems (MPS), consisting of interacting organs-on-chips or tissue-engineered, 3D organ constructs that use human cells, present an opportunity to bring new tools to biology, medicine, pharmacology, physiology, and toxicology. This issue of Experimental Biology and Medicine describes the ongoing development of MPS that can serve as in-vitro models for bone and cartilage, brain, gastrointestinal tract, lung, liver, microvasculature, reproductive tract, skeletal muscle, and skin. Related topics addressed here are the interconnection of organs-on-chips to support physiologically based pharmacokinetics and drug discovery and screening, and the microscale technologies that regulate stem cell differentiation. The initial motivation for creating MPS was to increase the speed, efficiency, and safety of pharmaceutical development and testing, paying particular regard to the fact that neither monolayer monocultures of immortal or primary cell lines nor animal studies can adequately recapitulate the dynamics of drug-organ, drug-drug, and drug-organ-organ interactions in humans. Other applications include studies of the effect of environmental toxins on humans, identification, characterization, and neutralization of chemical and biological weapons, controlled studies of the microbiome and infectious disease that cannot be conducted in humans, controlled differentiation of induced pluripotent stem cells into specific adult cellular phenotypes, and studies of the dynamics of metabolism and signaling within and between human organs. The technical challenges are being addressed by many investigators, and in the process, it seems highly likely that significant progress will be made toward providing more physiologically realistic alternatives to monolayer monocultures or whole animal studies. The effectiveness of this effort will be determined in part by how easy the constructs are to use, how well they function, how accurately they recapitulate and report human pharmacology and toxicology, whether they can be generated in large numbers to enable parallel studies, and if their use can be standardized consistent with the practices of regulatory science.
Collapse
Affiliation(s)
- John P Wikswo
- Departments of Biomedical Engineering, Molecular Physiology and Biophysics, and Physics and Astronomy, Vanderbilt University, The Vanderbilt Institute for Integrative Biosystems Research and Education, VU Station B 351807, Nashville, TN 37235-1807, USA
| |
Collapse
|