1
|
R N, Aggarwal A, Sravani AB, Mallya P, Lewis S. Organ-On-A-Chip: An Emerging Research Platform. Organogenesis 2023; 19:2278236. [PMID: 37965897 PMCID: PMC10653779 DOI: 10.1080/15476278.2023.2278236] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
In drug development, conventional preclinical and clinical testing stages rely on cell cultures and animal experiments, but these methods may fall short of fully representing human biology. To overcome this limitation, the emergence of organ-on-a-chip (OOC) technology has sparked interest as a transformative approach in drug testing research. By closely replicating human organ responses to external signals, OOC devices hold immense potential in revolutionizing drug efficacy and safety predictions. This review focuses on the advancements, applications, and prospects of OOC devices in drug testing. Based on the latest advances in the field of OOC systems and their clinical applications, this review reflects the effectiveness of OOC devices in replacing human volunteers in certain clinical studies. This review underscores the critical role of OOC technology in transforming drug testing methodologies.
Collapse
Affiliation(s)
- Nithin R
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Ayushi Aggarwal
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Anne Boyina Sravani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Pooja Mallya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
2
|
Colombo R, Ferron L, Frosi I, Papetti A. Advances in static in vitro digestion models after the COST action Infogest consensus protocol. Food Funct 2021; 12:7619-7636. [PMID: 34250533 DOI: 10.1039/d1fo01089a] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In vitro digestion models are essential to predictively evaluate the bioaccessibility and bioactivity of food molecules or natural products. Dynamic models better simulate the gastrointestinal conditions as they reproduce similar physiological environments. Despite this, static methods, also known as biochemical methods, represent a simple and useful approach for the study of different types of molecules, with a broad applicability in the nutritional, pharmaceutical, and toxicological fields. In addition, static models can be validated, avoiding the disadvantage of a difficult reproducibility of dynamic in vitro systems and inter-individual variations of in vivo experiments. A crucial point in the standardization of static models was the COST Action Infogest in 2014, which elaborated an international consensus static digestion method to harmonize experimental conditions and has general guidelines, thus allowing the comparison of studies and data. The aim of our review is to underline the impact of the Infogest consensus method and the development and evolution of in vitro static methods in the following years, with a focus on food applications.
Collapse
Affiliation(s)
- Raffaella Colombo
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100, Pavia, Italy.
| | | | | | | |
Collapse
|
3
|
Cappellozza E, Zanzoni S, Malatesta M, Calderan L. Integrated Microscopy and Metabolomics to Test an Innovative Fluid Dynamic System for Skin Explants In Vitro. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2021; 27:923-934. [PMID: 34311807 DOI: 10.1017/s1431927621012010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The in vitro models are receiving growing attention in studies on skin permeation, penetration, and irritancy, especially for the preclinical development of new transcutaneous drugs. However, synthetic membranes or cell cultures are unable to effectively mimic the permeability and absorption features of the cutaneous barrier. The use of explanted skin samples maintained in a fluid dynamic environment would make it possible for an in vitro experimentation closer to in vivo physiological conditions. To this aim, in the present study, we have modified a bioreactor designed for cell culture to host explanted skin samples. The preservation of the skin was evaluated by combining light, transmission, and scanning electron microscopy, for the histo/cytological characterization, with nuclear magnetic resonance spectroscopy, for the identification in the culture medium of metabolites indicative of the functional state of the explants. Our morphological and metabolomics results demonstrated that fluid dynamic conditions ameliorate significantly the structural and functional preservation of skin explants in comparison with conventional culture conditions. Our in vitro system is, therefore, reliable to test novel therapeutic agents intended for transdermal administration in skin samples from biopsies or surgical materials, providing predictive information suitable for focused in vivo research and reducing animal experimentation.
Collapse
Affiliation(s)
- Enrica Cappellozza
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Anatomy and Histology, School of Medicine and Surgery, University of Verona, Verona37134, Italy
| | - Serena Zanzoni
- Centro Piattaforme Tecnologiche, Spectroscopy, Diffractometry and Molecular Interaction Study Platform, University of Verona, Verona37100, Italy
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Anatomy and Histology, School of Medicine and Surgery, University of Verona, Verona37134, Italy
| | - Laura Calderan
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Anatomy and Histology, School of Medicine and Surgery, University of Verona, Verona37134, Italy
| |
Collapse
|
4
|
Sung JH. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin Drug Metab Toxicol 2021; 17:969-986. [PMID: 33764248 DOI: 10.1080/17425255.2021.1908996] [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] [Indexed: 12/12/2022]
Abstract
Introduction: Accurate prediction of pharmacokinetic (PK) and toxicokinetics (TK) of drugs is imperative for successful development of new pharmaceutics. Although conventional in vitro methods for predicting the PK and TK of drugs are well established, limitations still exist and more advanced chip-based in vitro platforms combined with mathematical models can help researchers overcome the limitations. Areas covered: We will review recent progress in the development of multi-organ-on-a-chip platforms for predicting PK and TK of drugs, as well as mathematical approaches that can be combined with these platforms for experiment design, data analysis and in vitro-in vivo extrapolation (IVIVE) for application to humans. Expert opinion: Although there remain some challenges to be addressed, the remarkable progress in the area of multi-organ-on-a-chip in recent years indicate that we will see tangible outcomes that can be utilized in the pharmaceutical industry in near future.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, sejong, Republic of Korea
| |
Collapse
|
5
|
Colombo R, Paolillo M, Papetti A. A new millifluidic-based gastrointestinal platform to evaluate the effect of simulated dietary methylglyoxal intakes. Food Funct 2020; 10:4330-4338. [PMID: 31273366 DOI: 10.1039/c9fo00332k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The search for new in vitro modular bioreactors to simulate flow-mediated transport and absorption of chemical substances is a very important issue in toxicology and in drug and bioactive delivery research. The possibility of setting up a dynamic microenvironment leads to experimental conditions that may more closely resemble the in vivo model, especially to measure acute or chronic intake of compounds. We propose a novel millifluidic-based gastrointestinal model as an evolution of the common in vitro methods, to evaluate the exposure to exogenous methylglyoxal (MGO), a highly reactive α-oxoaldehyde responsible for the formation of advanced glycation end products involved in a number of chronic diseases. Gastric and intestinal cells were seeded into two different chambers, creating a multi-compartmental system where fluids dynamically interact with human gastric stromal and intestinal cells. MGO was tested at concentrations simulating different MGO food intakes (meal, daily, and hypothetically weekly). Cell viability was measured over time, and simultaneously, extracellular MGO was quantified by a validated RP-HPLC-DAD method to evaluate its absorption/metabolization. This new platform gives the opportunity to connect different compartments, allowing studying kinetic and metabolic profiles of different substances and representing a very promising alternative to animal models, at least in preliminary investigations.
Collapse
Affiliation(s)
- Raffaella Colombo
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100, Pavia, Italy.
| | | | | |
Collapse
|
6
|
Sung JH, Wang Y, Shuler ML. Strategies for using mathematical modeling approaches to design and interpret multi-organ microphysiological systems (MPS). APL Bioeng 2019; 3:021501. [PMID: 31263796 PMCID: PMC6586554 DOI: 10.1063/1.5097675] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Recent advances in organ-on-a-chip technology have resulted in numerous examples of microscale systems that faithfully mimic the physiology and pathology of human organs and diseases. The next step in this field, which has already been partially demonstrated at a proof-of-concept level, would be integration of organ modules to construct multiorgan microphysiological systems (MPSs). In particular, there is interest in "body-on-a-chip" models, which recapitulate complex and dynamic interactions between different organs. Integration of multiple organ modules, while faithfully reflecting human physiology in a quantitative sense, will require careful consideration of factors such as relative organ sizes, blood flow rates, cell numbers, and ratios of cell types. The use of a mathematical modeling platform will be an essential element in designing multiorgan MPSs and interpretation of experimental results. Also, extrapolation to in vivo will require robust mathematical modeling techniques. So far, several scaling methods and pharmacokinetic and physiologically based pharmacokinetic models have been applied to multiorgan MPSs, with each method being suitable to a subset of different objectives. Here, we summarize current mathematical methodologies used for the design and interpretation of multiorgan MPSs and suggest important considerations and approaches to allow multiorgan MPSs to recapitulate human physiology and disease progression better, as well as help in vitro to in vivo translation of studies on response to drugs or chemicals.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| | - Ying Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
7
|
A novel dynamic multicellular co-culture system for studying individual blood-brain barrier cell types in brain diseases and cytotoxicity testing. Sci Rep 2018; 8:8784. [PMID: 29884831 PMCID: PMC5993789 DOI: 10.1038/s41598-018-26480-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 05/08/2018] [Indexed: 01/10/2023] Open
Abstract
Blood brain barrier (BBB) cells play key roles in the physiology and pathology of the central nervous system (CNS). BBB dysfunction is implicated in many neurodegenerative diseases, including Alzheimer’s disease (AD). The BBB consists of capillary endothelial cells, pericytes encircling the endothelium and surrounding astrocytes extending their processes towards it. Although there have been many attempts to develop in vitro BBB models, the complex interaction between these cell types makes it extremely difficult to determine their individual contribution to neurotoxicity in vivo. Thus, we developed and optimised an in vitro multicellular co-culture model within the Kirkstall Quasi Vivo System. The main aim was to determine the optimal environment to culture human brain primary endothelial cells, pericytes and astrocytes whilst maintaining cellular communication without formation of a barrier in order to assess the contribution of each cell type to the overall response. As a proof of concept for the present system, the effects of amyloid-beta 25-35 peptide (Aβ25-35), a hallmark of AD, were explored. This multicellular system will be a valuable tool for future studies on the specific roles of individual BBB cell type (while making connection with each other through medium) in CNS disorders as well as in cytotoxicity tests.
Collapse
|
8
|
Systemic and vascular inflammation in an in-vitro model of central obesity. PLoS One 2018; 13:e0192824. [PMID: 29438401 PMCID: PMC5811040 DOI: 10.1371/journal.pone.0192824] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Metabolic disorders due to over-nutrition are a major global health problem, often associated with obesity and related morbidities. Obesity is peculiar to humans, as it is associated with lifestyle and diet, and so difficult to reproduce in animal models. Here we describe a model of human central adiposity based on a 3-tissue system consisting of a series of interconnected fluidic modules. Given the causal link between obesity and systemic inflammation, we focused primarily on pro-inflammatory markers, examining the similarities and differences between the 3-tissue model and evidence from human studies in the literature. When challenged with high levels of adiposity, the in-vitro system manifests cardiovascular stress through expression of E-selectin and von Willebrand factor as well as systemic inflammation (expressing IL-6 and MCP-1) as observed in humans. Interestingly, most of the responses are dependent on the synergic interaction between adiposity and the presence of multiple tissue types. The set-up has the potential to reduce animal experiments in obesity research and may help unravel specific cellular mechanisms which underlie tissue response to nutritional overload.
Collapse
|
9
|
Carton F, Calderan L, Malatesta M. Incubation under fluid dynamic conditions markedly improves the structural preservation in vitro of explanted skeletal muscles. Eur J Histochem 2017; 61:2862. [PMID: 29313601 PMCID: PMC5745380 DOI: 10.4081/ejh.2017.2862] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 11/23/2022] Open
Abstract
Explanted organs and tissues represent suitable experimental systems mimicking the functional and structural complexity of the living organism, with positive ethical and economic impact on research activities. However, their preservation in culture is generally limited, thus hindering their application as experimental models for biomedical research. In the present study, we investigated the potential of an innovative fluid dynamic culture system to improve the structural preservation in vitro of explanted mouse skeletal muscles (soleus). We used light and transmission electron microscopy to compare the morphological features of muscles maintained either in multiwell plates under conventional conditions or in a bioreactor mimicking the flow of physiological fluids. Our results demonstrate that fluid dynamic conditions markedly slowed the progressive structural deterioration of the muscle tissue occurring during the permanence in the culture medium, prolonging the preservation of some organelles such as mitochondria up to 48 h.
Collapse
Affiliation(s)
- Flavia Carton
- University of Verona, Department of Neurological, Biomedical and Movement Sciences.
| | | | | |
Collapse
|
10
|
|
11
|
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
|
12
|
Lee SH, Ha SK, Choi I, Choi N, Park TH, Sung JH. Microtechnology-based organ systems and whole-body models for drug screening. Biotechnol J 2016; 11:746-56. [PMID: 27125245 DOI: 10.1002/biot.201500551] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/16/2016] [Accepted: 04/06/2016] [Indexed: 01/09/2023]
Abstract
After drug administration, the drugs are absorbed, distributed, metabolized, and excreted (ADME). Because ADME processes affect drug efficacy, various in vitro models have been developed based on the ADME processes. Although these models have been widely accepted as a tool for predicting the effects of drugs, the differences between in vivo and in vitro systems result in high attrition rates of drugs during the development process and remain a major limitation. Recent advances in microtechnology enable more accurate mimicking of the in vivo environment, where cellular behavior and physiological responses to drugs are more realistic; this has led to the development of novel in vitro systems, known as "organ-on-a-chip" systems. The development of organ-on-a-chip systems has progressed to include the reproduction of multiple organ interactions, which is an important step towards "body-on-a-chip" systems that will ultimately predict whole-body responses to drugs. In this review, we summarize the application of microtechnology for the development of in vitro systems that accurately mimic in vivo environments and reconstruct multiple organ models.
Collapse
Affiliation(s)
- Seung Hwan Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Sang Keun Ha
- Korea Food Research Institute, Seongnam, Gyeonggi-do, Republic of Korea
| | - Inwook Choi
- Korea Food Research Institute, Seongnam, Gyeonggi-do, Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea.,Advanced Institutes of Convergence Technology, Suwon, Gyeonggi-do, Republic of Korea
| | - Jong Hwan Sung
- Chemical Engineering, Hongik University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Usta OB, McCarty WJ, Bale S, Hegde M, Jindal R, Bhushan A, Golberg I, Yarmush ML. Microengineered cell and tissue systems for drug screening and toxicology applications: Evolution of in-vitro liver technologies. TECHNOLOGY 2015; 3:1-26. [PMID: 26167518 PMCID: PMC4494128 DOI: 10.1142/s2339547815300012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The liver performs many key functions, the most prominent of which is serving as the metabolic hub of the body. For this reason, the liver is the focal point of many investigations aimed at understanding an organism's toxicological response to endogenous and exogenous challenges. Because so many drug failures have involved direct liver toxicity or other organ toxicity from liver generated metabolites, the pharmaceutical industry has constantly sought superior, predictive in-vitro models that can more quickly and efficiently identify problematic drug candidates before they incur major development costs, and certainly before they are released to the public. In this broad review, we present a survey and critical comparison of in-vitro liver technologies along a broad spectrum, but focus on the current renewed push to develop "organs-on-a-chip". One prominent set of conclusions from this review is that while a large body of recent work has steered the field towards an ever more comprehensive understanding of what is needed, the field remains in great need of several key advances, including establishment of standard characterization methods, enhanced technologies that mimic the in-vivo cellular environment, and better computational approaches to bridge the gap between the in-vitro and in-vivo results.
Collapse
Affiliation(s)
- O B Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - W J McCarty
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - S Bale
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - M Hegde
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - R Jindal
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - A Bhushan
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - I Golberg
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - M L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA ; Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
14
|
Ucciferri N, Sbrana T, Ahluwalia A. Allometric Scaling and Cell Ratios in Multi-Organ in vitro Models of Human Metabolism. Front Bioeng Biotechnol 2014; 2:74. [PMID: 25566537 PMCID: PMC4269269 DOI: 10.3389/fbioe.2014.00074] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/04/2014] [Indexed: 11/13/2022] Open
Abstract
Intelligent in vitro models able to recapitulate the physiological interactions between tissues in the body have enormous potential as they enable detailed studies on specific two-way or higher order tissue communication. These models are the first step toward building an integrated picture of systemic metabolism and signaling in physiological or pathological conditions. However, the rational design of in vitro models of cell–cell or cell–tissue interaction is difficult as quite often cell culture experiments are driven by the device used, rather than by design considerations. Indeed, very little research has been carried out on in vitro models of metabolism connecting different cell or tissue types in a physiologically and metabolically relevant manner. Here, we analyze the physiological relationship between cells, cell metabolism, and exchange in the human body using allometric rules, downscaling them to an organ-on-a-plate device. In particular, in order to establish appropriate cell ratios in the system in a rational manner, two different allometric scaling models (cell number scaling model and metabolic and surface scaling model) are proposed and applied to a two compartment model of hepatic-vascular metabolic cross-talk. The theoretical scaling studies illustrate that the design and hence relevance of multi-organ models is principally determined by experimental constraints. Two experimentally feasible model configurations are then implemented in a multi-compartment organ-on-a-plate device. An analysis of the metabolic response of the two configurations demonstrates that their glucose and lipid balance is quite different, with only one of the two models recapitulating physiological-like homeostasis. In conclusion, not only do cross-talk and physical stimuli play an important role in in vitro models, but the numeric relationship between cells is also crucial to recreate in vitro interactions, which can be extrapolated to the in vivo reality.
Collapse
Affiliation(s)
- Nadia Ucciferri
- CNR Institute of Clinical Physiology , Pisa , Italy ; Interdepartmental Research Center "E. Piaggio", University of Pisa , Pisa , Italy
| | - Tommaso Sbrana
- Interdepartmental Research Center "E. Piaggio", University of Pisa , Pisa , Italy
| | - Arti Ahluwalia
- CNR Institute of Clinical Physiology , Pisa , Italy ; Interdepartmental Research Center "E. Piaggio", University of Pisa , Pisa , Italy
| |
Collapse
|
15
|
Andreoni C, Orsi G, De Maria C, Montemurro F, Vozzi G. In silico models for dynamic connected cell cultures mimicking hepatocyte-endothelial cell-adipocyte interaction circle. PLoS One 2014; 9:e111946. [PMID: 25502576 PMCID: PMC4266517 DOI: 10.1371/journal.pone.0111946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 10/09/2014] [Indexed: 01/12/2023] Open
Abstract
The biochemistry of a system made up of three kinds of cell is virtually impossible to work out without the use of in silico models. Here, we deal with homeostatic balance phenomena from a metabolic point of view and we present a new computational model merging three single-cell models, already available from our research group: the first model reproduced the metabolic behaviour of a hepatocyte, the second one represented an endothelial cell, and the third one described an adipocyte. Multiple interconnections were created among these three models in order to mimic the main physiological interactions that are known for the examined cell phenotypes. The ultimate aim was to recreate the accomplishment of the homeostatic balance as it was observed for an in vitro connected three-culture system concerning glucose and lipid metabolism in the presence of the medium flow. The whole model was based on a modular approach and on a set of nonlinear differential equations implemented in Simulink, applying Michaelis-Menten kinetic laws and some energy balance considerations to the studied metabolic pathways. Our in silico model was then validated against experimental datasets coming from literature about the cited in vitro model. The agreement between simulated and experimental results was good and the behaviour of the connected culture system was reproduced through an adequate parameter evaluation. The developed model may help other researchers to investigate further about integrated metabolism and the regulation mechanisms underlying the physiological homeostasis.
Collapse
Affiliation(s)
- Chiara Andreoni
- Research Center “E. Piaggio”, University of Pisa, Pisa, Italy
- * E-mail:
| | - Gianni Orsi
- Research Center “E. Piaggio”, University of Pisa, Pisa, Italy
| | - Carmelo De Maria
- Research Center “E. Piaggio”, University of Pisa, Pisa, Italy
- Department of Information Engineering, University of Pisa, Pisa, Italy
| | | | - Giovanni Vozzi
- Research Center “E. Piaggio”, University of Pisa, Pisa, Italy
- Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
16
|
Design Criteria for Generating Physiologically Relevant In Vitro Models in Bioreactors. Processes (Basel) 2014. [DOI: 10.3390/pr2030548] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
17
|
Iori E, Vinci B, Murphy E, Marescotti MC, Avogaro A, Ahluwalia A. Glucose and fatty acid metabolism in a 3 tissue in-vitro model challenged with normo- and hyperglycaemia. PLoS One 2012; 7:e34704. [PMID: 22509346 PMCID: PMC3324505 DOI: 10.1371/journal.pone.0034704] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/08/2012] [Indexed: 11/18/2022] Open
Abstract
Nutrient balance in the human body is maintained through systemic signaling between different cells and tissues. Breaking down this circuitry to its most basic elements and reconstructing the metabolic network in-vitro provides a systematic method to gain a better understanding of how cross-talk between the organs contributes to the whole body metabolic profile and of the specific role of each different cell type. To this end, a 3-way connected culture of hepatocytes, adipose tissue and endothelial cells representing a simplified model of energetic substrate metabolism in the visceral region was developed. The 3-way culture was shown to maintain glucose and fatty acid homeostasis in-vitro. Subsequently it was challenged with insulin and high glucose concentrations to simulate hyperglycaemia. The aim was to study the capacity of the 3-way culture to maintain or restore normal circulating glucose concentrations in response to insulin and to investigate the effects these conditions on other metabolites involved in glucose and lipid metabolism. The results show that the system’s metabolic profile changes dramatically in the presence of high concentrations of glucose, and that these changes are modulated by the presence of insulin. Furthermore, we observed an increase in E-selectin levels in hyperglycaemic conditions and increased IL-6 concentrations in insulin-free-hyperglycaemic conditions, indicating, respectively, endothelial injury and proinflammatory stress in the challenged 3-way system.
Collapse
Affiliation(s)
- Elisabetta Iori
- Division of Metabolic Diseases, Department of Clinical and Experimental Medicine, University of Padua, Padua, Italy
| | - Bruna Vinci
- Centro Interdipartimentale di Ricerca ″E.Piaggio″, University of Pisa, Pisa, Italy
- CNR Institute of Clinical Physiology, Pisa, Italy
| | - Ellen Murphy
- Division of Metabolic Diseases, Department of Clinical and Experimental Medicine, University of Padua, Padua, Italy
| | - Maria Cristina Marescotti
- Division of Metabolic Diseases, Department of Clinical and Experimental Medicine, University of Padua, Padua, Italy
| | - Angelo Avogaro
- Division of Metabolic Diseases, Department of Clinical and Experimental Medicine, University of Padua, Padua, Italy
| | - Arti Ahluwalia
- Centro Interdipartimentale di Ricerca ″E.Piaggio″, University of Pisa, Pisa, Italy
- CNR Institute of Clinical Physiology, Pisa, Italy
- * E-mail:
| |
Collapse
|
18
|
BiotecVisions 2012, February. Biotechnol J 2012. [DOI: 10.1002/biot.201100505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|