101
|
Boeri L, Izzo L, Sardelli L, Tunesi M, Albani D, Giordano C. Advanced Organ-on-a-Chip Devices to Investigate Liver Multi-Organ Communication: Focus on Gut, Microbiota and Brain. Bioengineering (Basel) 2019; 6:E91. [PMID: 31569428 PMCID: PMC6956143 DOI: 10.3390/bioengineering6040091] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023] Open
Abstract
The liver is a key organ that can communicate with many other districts of the human body. In the last few decades, much interest has focused on the interaction between the liver and the gut microbiota, with their reciprocal influence on biosynthesis pathways and the integrity the intestinal epithelial barrier. Dysbiosis or liver disorders lead to0 epithelial barrier dysfunction, altering membrane permeability to toxins. Clinical and experimental evidence shows that the permeability hence the delivery of neurotoxins such as LPS, ammonia and salsolinol contribute to neurological disorders. These findings suggested multi-organ communication between the gut microbiota, the liver and the brain. With a view to in vitro modeling this liver-based multi-organ communication, we describe the latest advanced liver-on-a-chip devices and discuss the need for new organ-on-a-chip platforms for in vitro modeling the in vivo multi-organ connection pathways in physiological and pathological situations.
Collapse
Affiliation(s)
- Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy.
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy.
| | - Lorenzo Sardelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy.
| | - Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy.
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milan, Italy.
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy.
| |
Collapse
|
102
|
Bulutoglu B, Rey-Bedón C, Kang YBA, Mert S, Yarmush ML, Usta OB. A microfluidic patterned model of non-alcoholic fatty liver disease: applications to disease progression and zonation. LAB ON A CHIP 2019; 19:3022-3031. [PMID: 31465069 PMCID: PMC6736752 DOI: 10.1039/c9lc00354a] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its progressive form non-alcoholic steatohepatitis (NASH) affect 25% of the world population. NAFLD is predicted to soon become the main cause of liver morbidity and transplantation. The disease is characterized by a progressive increase of lipid accumulation in hepatocytes, which eventually induce fibrosis and inflammation, and can ultimately cause cirrhosis and hepatic carcinoma. Here, we created a patterned model of NAFLD on a chip using free fatty acid gradients to recapitulate a spectrum of disease conditions in a single continuous liver tissue. We established the NAFLD progression via quantification of intracellular lipid accumulation and transcriptional levels of fatty acid transporters and NAFLD pathogenesis markers. We then used this platform to create oxygen driven steatosis zonation mimicking the sinusoidal lipid distribution on a single continuous tissue and showed that this fat zonation disappears under progressed steatosis, in agreement with in vivo observations and recent computational studies. While we focus on free fatty acids and oxygen as the drivers of NAFLD, the microfluidic platform here is extensible to simultaneous use of other drivers.
Collapse
Affiliation(s)
- Beyza Bulutoglu
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA 02114, USA.
| | | | | | | | | | | |
Collapse
|
103
|
Müller FA, Sturla SJ. Human in vitro models of nonalcoholic fatty liver disease. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.03.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
104
|
Lasli S, Kim HJ, Lee K, Suurmond CAE, Goudie M, Bandaru P, Sun W, Zhang S, Zhang N, Ahadian S, Dokmeci MR, Lee J, Khademhosseini A. A Human Liver-on-a-Chip Platform for Modeling Nonalcoholic Fatty Liver Disease. ADVANCED BIOSYSTEMS 2019; 3:e1900104. [PMID: 32648699 PMCID: PMC7473489 DOI: 10.1002/adbi.201900104] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/29/2019] [Indexed: 12/16/2022]
Abstract
The liver possesses a unique microenvironment with a complex internal vascular system and cell-cell interactions. Nonalcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease, and although much effort has been dedicated to building models to target NAFLD, most in vitro systems rely on simple models failing to recapitulate complex liver functions. Here, an in vitro system is presented to study NAFLD (steatosis) by coculturing human hepatocellular carcinoma (HepG2) cells and umbilical vein endothelial cells (HUVECs) into spheroids. Analysis of colocalization of HepG2-HUVECs along with the level of steatosis reveals that the NAFLD pathogenesis could be better modeled when 20% of HUVECs are presented in HepG2 spheroids. Spheroids with fat supplements progressed to the steatosis stage on day 2, which could be maintained for more than a week without being harmful for cells. Transferring spheroids onto a chip system with an array of interconnected hexagonal microwells proves helpful for monitoring functionality through increased albumin secretions with HepG2-HUVEC interactions and elevated production of reactive oxygen species for steatotic spheroids. The reversibility of steatosis is demonstrated by simply stopping fat-based diet or by antisteatotic drug administration, the latter showing a faster return of intracellular lipid levels to the basal level.
Collapse
Affiliation(s)
- Soufian Lasli
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Han-Jun Kim
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - KangJu Lee
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Ceri-Anne E Suurmond
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Bioengineering Technologies, University of Twente, 7522, NB, Enschede, The Netherlands
| | - Marcus Goudie
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Praveen Bandaru
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Wujin Sun
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Shiming Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Niyuan Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Samad Ahadian
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Mehmet R Dokmeci
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Junmin Lee
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Ali Khademhosseini
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
105
|
Park D, Lee J, Chung JJ, Jung Y, Kim SH. Integrating Organs-on-Chips: Multiplexing, Scaling, Vascularization, and Innervation. Trends Biotechnol 2019; 38:99-112. [PMID: 31345572 DOI: 10.1016/j.tibtech.2019.06.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Organs-on-chips (OoCs) have attracted significant attention because they can be designed to mimic in vivo environments. Beyond constructing a single OoC, recent efforts have tried to integrate multiple OoCs to broaden potential applications such as disease modeling and drug discoveries. However, various challenges remain for integrating OoCs towards in vivo-like operation, such as incorporating various connections for integrating multiple OoCs. We review multiplexed OoCs and challenges they face: scaling, vascularization, and innervation. In our opinion, future OoCs will be constructed to have increased predictive power for in vivo phenomena and will ultimately become a mainstream tool for high quality biomedical and pharmaceutical research.
Collapse
Affiliation(s)
- DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jaeseo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Justin J Chung
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Biomaterials Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
106
|
Morss Clyne A, Swaminathan S, Díaz Lantada A. Biofabrication strategies for creating microvascular complexity. Biofabrication 2019; 11:032001. [PMID: 30743247 DOI: 10.1088/1758-5090/ab0621] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Design and fabrication of effective biomimetic vasculatures constitutes a relevant and yet unsolved challenge, lying at the heart of tissue repair and regeneration strategies. Even if cell growth is achieved in 3D tissue scaffolds or advanced implants, tissue viability inevitably requires vascularization, as diffusion can only transport nutrients and eliminate debris within a few hundred microns. This engineered vasculature may need to mimic the intricate branching geometry of native microvasculature, referred to herein as vascular complexity, to efficiently deliver blood and recreate critical interactions between the vascular and perivascular cells as well as parenchymal tissues. This review first describes the importance of vascular complexity in labs- and organs-on-chips, the biomechanical and biochemical signals needed to create and maintain a complex vasculature, and the limitations of current 2D, 2.5D, and 3D culture systems in recreating vascular complexity. We then critically review available strategies for design and biofabrication of complex vasculatures in cell culture platforms, labs- and organs-on-chips, and tissue engineering scaffolds, highlighting their advantages and disadvantages. Finally, challenges and future directions are outlined with the hope of inspiring researchers to create the reliable, efficient and sustainable tools needed for design and biofabrication of complex vasculatures.
Collapse
Affiliation(s)
- Alisa Morss Clyne
- Vascular Kinetics Laboratory, Mechanical Engineering & Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, United States of America
| | | | | |
Collapse
|
107
|
Cedillo-Alcantar DF, Han YD, Choi J, Garcia-Cordero JL, Revzin A. Automated Droplet-Based Microfluidic Platform for Multiplexed Analysis of Biochemical Markers in Small Volumes. Anal Chem 2019; 91:5133-5141. [PMID: 30834743 DOI: 10.1021/acs.analchem.8b05689] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The ability to detect multiple analytes in a small sample volume has significance for numerous areas of research, including organs-on-chip, small animal experiments, and neonatology. The objective of this study was to develop an automated microfluidics platform for multiplexed detection of analytes in microliter sample volumes. This platform employed computer-controlled microvalves to create laminar co-flows of sample and assay reagent solutions. It also contained valve-regulated cross-junction for discretizing sample/reagent mixtures into water-in-oil droplets. Microfluidic automation allowed us to control parameters related to frequency of droplet generation and the number of droplets of the same composition, as well as the size of droplets. Each droplet represented an individual enzymatic assay carried out in a sub-nanoliter (0.8 nL) volume reactor. An enzymatic reaction involving target analyte and assay reagents produced colorimetric or fluorescent signals in droplets. Importantly, intensity of optical signal was proportional to the concentration of analyte in question. This microfluidic bioanalysis platform was used in conjunction with commercial "mix-detect" assays for glucose, total bile acids, and lactate dehydrogenase (LDH). After characterizing these assays individually, we demonstrated sensitive multiplexed detection of three analytes from as little as 3 μL. In fact, this volume was sufficient to generate multiple repeat droplets for each of the three biochemical assays as well as positive control droplets, confirming the quality of assay reagents and negative control droplets to help with background subtraction. One potential application for this microfluidic bioanalysis platform involves sampling cell-conditioned media in organ-on-chip devices. To highlight this application, hepatocyte spheroids were established in microfluidic devices, injured on-chip by exposure to lipotoxic agent (palmitate), and then connected to the bioanalysis module for daily monitoring of changes in cytotoxicity (LDH), energy metabolism (glucose), and liver function (total bile acids). Microfluidic in-droplet assays revealed increased levels of LDH as well as reduction in bile acid synthesis-results that were consistent with hepatic injury. Importantly, these experiments highlighted the fact that in-droplet assays were sufficiently sensitive to detect changes in functional output of a relatively small (∼100) number of hepatocyte spheroids cultured in a microfluidic device. Moving forward, we foresee increasing the multiplexing capability of this technology and applying this platform to other biological/medical scenarios where detection of multiple analytes from a small sample volume is desired.
Collapse
Affiliation(s)
- Diana F Cedillo-Alcantar
- Unidad Monterrey , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Vía del Conocimiento 201 , Parque PIIT, Apodaca , Nuevo León CP 66628 , México.,Mayo Clinic , Rochester , Minnesota 55905 , United States
| | - Yong Duk Han
- Mayo Clinic , Rochester , Minnesota 55905 , United States
| | - Jonghoon Choi
- Mayo Clinic , Rochester , Minnesota 55905 , United States
| | - Jose L Garcia-Cordero
- Unidad Monterrey , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Vía del Conocimiento 201 , Parque PIIT, Apodaca , Nuevo León CP 66628 , México
| | | |
Collapse
|
108
|
Sivakumar P, Kitson C, Jarai G. Modeling and measuring extracellular matrix alterations in fibrosis: challenges and perspectives for antifibrotic drug discovery. Connect Tissue Res 2019; 60:62-70. [PMID: 30071759 DOI: 10.1080/03008207.2018.1500557] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
An imbalance of extracellular matrix (ECM) deposition and turnover is a hallmark of fibrotic pathologies as opposed to normal repair response to injury across several organs. Antifibrotic approaches to date have targeted multiple mechanisms and pathways involved in inflammation, angiogenesis, injury, wound repair, ECM biosynthesis, assembly, crosslinking and degradation. Many of these approaches have been unsuccessful which may in part be due to suboptimal models and the lack of validated functional ECM end points relevant to fibrosis. In addition, drug discovery and development for fibrotic diseases has been challenging due to the lack of translatability from in vivo models to the clinic. Targeting growth factor signaling pathways such as transforming growth factor beta (TGFβ), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF) are possible in simple recombinant cell models and the approval of the tyrosine kinase inhibitor, nintedanib (Ofev) is testament to the approach. However, drug targets directly impacting ECM synthesis, assembly or degradation have proven clinically intractable to date. The reasons for a lack of progress are many and include; non-traditional drug targets, lack of suitable high throughput screening assays and translational models, incomplete understanding of the role of the target. Here, we review the role of ECM in fibrosis, the challenges of ECM-targeted antifibrotic approaches, progress in the development of functional and biomarker-related ECM assays and where new translational models of fibrotic ECM remodeling could support drug discovery for fibrotic diseases.
Collapse
Affiliation(s)
- Pitchumani Sivakumar
- a Fibrosis Translational Research and Development , Bristol-Myers Squibb , Pennington , NJ , USA
| | - Christopher Kitson
- b Fibrosis Discovery Biology , Bristol-Myers Squibb , Pennington , NJ , USA
| | - Gabor Jarai
- a Fibrosis Translational Research and Development , Bristol-Myers Squibb , Pennington , NJ , USA
| |
Collapse
|
109
|
Shapshak P, Balaji S, Kangueane P, Chiappelli F, Somboonwit C, Menezes LJ, Sinnott JT. Innovative Technologies for Advancement of WHO Risk Group 4 Pathogens Research. GLOBAL VIROLOGY III: VIROLOGY IN THE 21ST CENTURY 2019. [PMCID: PMC7122670 DOI: 10.1007/978-3-030-29022-1_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Paul Shapshak
- Department of Internal Medicine, University of South Florida, Tampa, FL USA
| | - Seetharaman Balaji
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka India
| | | | - Francesco Chiappelli
- Oral Biology and Medicine, CHS 63-090, UCLA School of Dentistry Oral Biology and Medicine, CHS 63-090, Los Angeles, CA USA
| | | | - Lynette J. Menezes
- Department of Internal Medicine, University of South Florida, Tampa, FL USA
| | - John T. Sinnott
- Department of Internal Medicine, University of South Florida, Tampa, FL USA
| |
Collapse
|
110
|
Abstract
In native tissues, various cell types organize and spatiotemporally function and communicate with neighboring or remote cells in a highly regulated way. How can we replicate these amazing functional structures in vitro? From the view of a chemist, the heterogeneous cells and extracellular matrix (ECM) could be regarded as various chemical substrate materials for "synthetic" reactions during tissue engineering. But how can we accelerate these reactions? Microfluidics provides ideal solutions. Microfluidics could be metaphorically regarded as a miniature "biofactory", whereas the on-chip critical chemical cues such as biomolecule gradients and physical cues such as geometrical confinement, topological guidance, and mechanical stimulations, along with the external stimulations such as light, electricity, acoustics, and magnetics, could be regarded as "catalytic cues" which can accelerate the "synthetic reactions" by precisely and effectively manipulating a series of cell behaviors including cell adhesion, migration, growth, proliferation, differentiation, cell-cell interaction, and cell-matrix interaction to reduce activation energy of the "synthetic reactions". Thus, on the microfluidics platform, the "biofactory", various "synthetic" reactions take place to change the substrate materials (cells and ECM) into products (tissues) in a nonlinear way, which is a typical feature of a biological process. By precisely organizing the substrate materials and spatiotemporally controlling the activity of the products, as a "biofactory", the microfluidics system can not only "synthesize" living tissues but also recreate physiological or pathophysiological processes such as immune responses, angiogenesis, wound healing, and tumor metastasis in vitro to bring insights into the mechanisms underlying these processes taking place in vivo. In this Account, we borrow the concept of chemical "synthesis" to describe how to "synthesize" artificial tissues using microfluidics from a chemist's view. Accelerated by the built-in physiochemical cues on microfluidics and external stimulations, various tissues could be "synthesized" on a microfluidics platform. We summarize that there are "step-by-step synthesis" and "one-step synthesis" on microfluidics for creating desired tissues with unprecedented precision, accuracy, and speed. In recent years, researchers developed various microfluidic techniques including creating adhesive domains for mediating reverse and precise adhesion, chemical gradients for directing cell growth, geometrical confinements and topological cues for manipulating cell migration, and mechanics for stimulating cell differentiation. By employing and orchestrating these on-chip tissue "synthetic" conditions, "step-by-step synthesis" could be realized on chips to develop multilayered tissues such as blood vessels. "One-step synthesis" on chips could develop functional three-dimensional tissue structures such as neural networks or nephron-like structures. Based on these on-chip studies, many critical physiological and pathophysiological processes such as wound healing, tumor metastasis, and atherosclerosis could be deeply investigated, and the drugs or therapeutic approaches could also be evaluated or screened conveniently. The "synthetic tissues on microfluidics" system would pave an avenue for precise creation of artificial tissues for not only fundamental research but also biomedical applications such as tissue engineering.
Collapse
Affiliation(s)
- Wenfu Zheng
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, P. R. China
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, P. R. China
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
- The University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
111
|
Human hepatic 3D spheroids as a model for steatosis and insulin resistance. Sci Rep 2018; 8:14297. [PMID: 30250238 PMCID: PMC6155201 DOI: 10.1038/s41598-018-32722-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a public health concern as reflected in its widespread distribution in the general population. Yet, treatment options are scarce which is at least in part due to lack of reliable human in vitro disease models. Here, we report a human hepatic 3D spheroid system cultured under defined chemical conditions that has the potential to mimic steatotic conditions in a reversible manner, useful for identification of novel drug treatment conditions. Primary human hepatocytes (PHH) from different donors were cultured as spheroid microtissues in physiological in vivo -like culture conditions. Hepatic steatosis was induced over the course of three weeks in culture by supplementing the culture medium with pathophysiological concentrations of free fatty acids, carbohydrates and insulin. Effects of steatosis in the 3D system were evaluated on transcriptional, metabolomic and lipidomic levels. Free fatty acids on one hand as well as a combination of insulin and monosaccharides, promoted lipid accumulation in hepatocytes and increased expression of lipogenic genes, such as fatty acid synthase. This milieu also promoted development of insulin resistance within 2 weeks as manifested by an increase in gluconeogenic and insulin resistance markers, which are observed in type 2 diabetes mellitus and metabolic syndrome. Induced steatosis was reversible after withdrawal of lipogenic substrates and a further reduction in cellular fat content was observed following treatment with different antisteatotic compounds, such as metformin, glucagon, olaparib and antioxidants. Taken together, these results demonstrate that the 3D hepatic spheroids can serve as a valuable, HTS compatible model for the study of liver steatosis and facilitate translational discovery of novel drug targets.
Collapse
|
112
|
Kulthong K, Duivenvoorde L, Mizera BZ, Rijkers D, Dam GT, Oegema G, Puzyn T, Bouwmeester H, van der Zande M. Implementation of a dynamic intestinal gut-on-a-chip barrier model for transport studies of lipophilic dioxin congeners. RSC Adv 2018; 8:32440-32453. [PMID: 35547722 PMCID: PMC9086222 DOI: 10.1039/c8ra05430d] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/10/2018] [Indexed: 11/21/2022] Open
Abstract
Novel microfluidic technologies allow the manufacture of in vitro organ-on-a-chip systems that hold great promise to adequately recapitulate the biophysical and functional complexity of organs found in vivo. In this study, a gut-on-a-chip model was developed aiming to study the potential cellular association and transport of food contaminants. Intestinal epithelial cells (Caco-2) were cultured on a porous polyester membrane that was tightly clamped between two glass slides to form two separate flow chambers. Glass syringes, polytetrafluoroethylene tubing and glass microfluidic chips were selected to minimize surface adsorption of the studied compounds (i.e. highly lipophilic dioxins), during the transport studies. Confocal microscopy studies revealed that, upon culturing under constant flow for 7 days, Caco-2 cells formed complete and polarized monolayers as observed after culturing for 21 days under static conditions in Transwells. We exposed Caco-2 monolayers in the chip and Transwell to a mixture of 17 dioxin congeners (7 polychlorinated dibenzo-p-dioxins and 10 polychlorinated dibenzofurans) for 24 h. Gas chromatography-high resolution mass spectrometry was used to assess the cellular association and transport of individual dioxin congeners across the Caco-2 cell monolayers. After 24 h, the amount of transported dioxin mixture was similar in both the dynamic gut-on-a-chip model and the static Transwell model. The transport of individual congeners corresponded with their number of chlorine atoms and substitution patterns as revealed by quantitative structure-property relationship modelling. These results show that the gut-on-a-chip model can be used, as well as the traditional static Transwell system, to study the cellular association and transport of lipophilic compounds like dioxins.
Collapse
Affiliation(s)
- Kornphimol Kulthong
- Division of Toxicology, Wageningen University P.O. Box 8000, 6700 EA, Wageningen The Netherlands
- RIKILT-Wageningen Research P.O. Box 230, 6700 AE, Wageningen The Netherlands
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Loes Duivenvoorde
- RIKILT-Wageningen Research P.O. Box 230, 6700 AE, Wageningen The Netherlands
| | - Barbara Z Mizera
- Laboratory of Environmental Chemometrics, Faculty of Chemistry, University of Gdansk Gdansk Poland
| | - Deborah Rijkers
- RIKILT-Wageningen Research P.O. Box 230, 6700 AE, Wageningen The Netherlands
| | - Guillaume Ten Dam
- RIKILT-Wageningen Research P.O. Box 230, 6700 AE, Wageningen The Netherlands
| | - Gerlof Oegema
- RIKILT-Wageningen Research P.O. Box 230, 6700 AE, Wageningen The Netherlands
| | - Tomasz Puzyn
- Laboratory of Environmental Chemometrics, Faculty of Chemistry, University of Gdansk Gdansk Poland
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University P.O. Box 8000, 6700 EA, Wageningen The Netherlands
| | - Meike van der Zande
- RIKILT-Wageningen Research P.O. Box 230, 6700 AE, Wageningen The Netherlands
| |
Collapse
|
113
|
Lee SY, Sung JH. Gut-liver on a chip toward an in vitro model of hepatic steatosis. Biotechnol Bioeng 2018; 115:2817-2827. [PMID: 29981260 DOI: 10.1002/bit.26793] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/31/2018] [Accepted: 07/02/2018] [Indexed: 12/16/2022]
Abstract
Hepatic steatosis is a process of abnormal lipid deposition within the liver cells, often caused by excessive alcohol uptake or obesity. A conventional in vitro model for hepatic steatosis uses a liver cell culture, treated with fatty acids and measures accumulation of lipids within the cells. This model does not recapitulate the complex process of absorption and metabolism of digestive lipids. Here, we introduce a gut-liver chip, which mimics the gut absorption and hepatic metabolism in a microfluidic chip. Absorption of fatty acids through gut layer and subsequent deposition within liver cells was demonstrated. Tumor necrosis factor-α, butyrate, and α-lipoic acid were chosen as model molecules that can affect hepatic steatosis via different mechanisms, and their effects were evaluated. Our results suggest that the gut-liver chip can mimic the absorption and accumulation of fatty acids in the gut and the liver.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Chemical Engineering, Hongik University, Seoul, Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, Korea
| |
Collapse
|
114
|
Li X, George SM, Vernetti L, Gough AH, Taylor DL. A glass-based, continuously zonated and vascularized human liver acinus microphysiological system (vLAMPS) designed for experimental modeling of diseases and ADME/TOX. LAB ON A CHIP 2018; 18:2614-2631. [PMID: 30063238 PMCID: PMC6113686 DOI: 10.1039/c8lc00418h] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The vLAMPS is a human, biomimetic liver MPS, in which the ECM and cell seeding of the intermediate layer prior to assembly, simplifies construction of the model and makes the platform user-friendly. This primarily glass microfluidic device is optimal for real-time imaging, while minimizing the binding of hydrophobic drugs/biologics to the materials that constitute the device. The assembly of the three layered device with primary human hepatocytes and liver sinusoidal endothelial cells (LSECs), and human cell lines for stellate and Kupffer cells, creates a vascular channel separated from the hepatic channel (chamber) by a porous membrane that allows communication between channels, recapitulating the 3D structure of the liver acinus. The vascular channel can be used to deliver drugs, immune cells, as well as various circulating cells and other factors to a stand-alone liver MPS and/or to couple the liver MPS to other organ MPS. We have successfully created continuous oxygen zonation by controlling the flow rates of media in the distinct vascular and hepatic channels and validated the computational modeling of zonation with oxygen sensitive and insensitive beads. This allows the direct investigation of the role of zonation in physiology, toxicology and disease progression. The vascular channel is lined with human LSECs, recapitulating partial immunologic functions within the liver sinusoid, including the activation of LSECs, promoting the binding of polymorphonuclear leukocytes (PMNs) followed by transmigration into the hepatic chamber. The vLAMPS is a valuable platform to investigate the functions of the healthy and diseased human liver using all primary human cell types and/or iPSC-derived cells.
Collapse
Affiliation(s)
- Xiang Li
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | | | | | | | | |
Collapse
|
115
|
Piemonte V, Cerbelli S, Capocelli M, Di Paola L, Prisciandaro M, Basile A. Design of microfluidic bioreactors: Transport regimes. ASIA-PAC J CHEM ENG 2018. [DOI: 10.1002/apj.2238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Vincenzo Piemonte
- Faculty of Engineering; University Campus Bio-Medico of Rome; Rome Italy
| | - Stefano Cerbelli
- Department of Chemical Engineering; University of Rome “La Sapienza”; Rome Italy
| | - Mauro Capocelli
- Faculty of Engineering; University Campus Bio-Medico of Rome; Rome Italy
| | - Luisa Di Paola
- Faculty of Engineering; University Campus Bio-Medico of Rome; Rome Italy
| | - Marina Prisciandaro
- Department of Industrial and Information Engineering and of Economics; University of L'Aquila; L'Aquila Italy
| | | |
Collapse
|
116
|
Yesil-Celiktas O, Hassan S, Miri AK, Maharjan S, Al-kharboosh R, Quiñones-Hinojosa A, Zhang YS. Mimicking Human Pathophysiology in Organ-on-Chip Devices. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800109] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ozlem Yesil-Celiktas
- Division of Engineering in Medicine; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Department of Bioengineering; Faculty of Engineering; Ege University; Bornova-Izmir 35100 Turkey
| | - Shabir Hassan
- Division of Engineering in Medicine; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
| | - Amir K. Miri
- Division of Engineering in Medicine; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Department of Mechanical Engineering Rowan University; 401 North Campus Drive Glassboro NJ 08028 USA
| | - Sushila Maharjan
- Division of Engineering in Medicine; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Research Institute for Bioscience and Biotechnology; Nakkhu-4 Lalitpur 44600 Nepal
| | - Rawan Al-kharboosh
- Mayo Clinic College of Medicine; Mayo Clinic Graduate School; Neuroscience, NBD Track Rochester MN 55905 USA
- Department of Neurosurgery, Oncology, Neuroscience; Mayo Clinic; Jacksonville FL 32224 USA
| | | | - Yu Shrike Zhang
- Division of Engineering in Medicine; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
| |
Collapse
|
117
|
Boeckmans J, Natale A, Buyl K, Rogiers V, De Kock J, Vanhaecke T, Rodrigues RM. Human-based systems: Mechanistic NASH modelling just around the corner? Pharmacol Res 2018; 134:257-267. [PMID: 29964161 DOI: 10.1016/j.phrs.2018.06.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease characterized by excessive triglyceride accumulation in the liver accompanied by inflammation, cell stress and apoptosis. It is the tipping point to the life-threatening stages of non-alcoholic fatty liver disease (NAFLD). Despite the high prevalence of NASH, up to five percent of the global population, there are currently no approved drugs to treat this disease. Animal models, mostly based on specific diets and genetic modifications, are often employed in anti-NASH drug development. However, due to interspecies differences and artificial pathogenic conditions, they do not represent the human situation accurately and are inadequate for testing the efficacy and safety of potential new drugs. Human-based in vitro models provide a more legitimate representation of the human NASH pathophysiology and can be used to investigate the dysregulation of cellular functions associated with the disease. Also in silico methodologies and pathway-based approaches using human datasets, may contribute to a more accurate representation of NASH, thereby facilitating the quest for new anti-NASH drugs. In this review, we describe the molecular components of NASH and how human-based tools can contribute to unraveling the pathogenesis of this disease and be used in anti-NASH drug development. We also propose a roadmap for the development and application of human-based approaches for future investigation of NASH.
Collapse
Affiliation(s)
- Joost Boeckmans
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Alessandra Natale
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Karolien Buyl
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Vera Rogiers
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Joery De Kock
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Tamara Vanhaecke
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Robim M Rodrigues
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
118
|
Cheng S, Yang Y, Zhou Y, Xiang W, Yao H, Ma L. Influence of different concentrations of uric acid on oxidative stress in steatosis hepatocytes. Exp Ther Med 2018; 15:3659-3665. [PMID: 29545896 PMCID: PMC5840957 DOI: 10.3892/etm.2018.5855] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
The development of nonalcoholic fatty liver disease (NAFLD) is caused by the steatosis of hepatocytes, which induces oxidative stress (OS). Thus, OS has an important role in the development of NAFLD. In the present study, the L-02 hepatocyte cell line was used to develop a steatosis cell model. The best model was determined using an MTT assay and the triglyceride levels. Model cells were treated with high concentrations of uric acid (UA; 0, 5, 10, 20 and 30 mg/dl) for 24, 48, 72 and 96 h. Indicators of oxidation were then measured, which included total superoxide dismutase (SOD), malonaldehyde (MDA) and reduced glutathione (GSH), and the transcriptional and translational levels of SOD1 and γ-glutamate-cysteine ligase (γ-GCLC) were also determined. In addition, the intracellular levels of aspartate aminotransferase and alanine aminotransferase (ALT) were detected. The activity of SOD1 decreased over time and the result was supported by the results of western blotting. The transcriptional levels of SOD1 in model cells was significantly higher than untreated cells at 48 h. With the decreased levels of SOD1 and GSH, MDA increased in a time-dependent manner. The content of GSH decreased with time as well, which was also reflected in the results of western blotting. The transcriptional levels of γ-GCLC in all UA-treated groups were lower when compared with those observed in the model group. The activity of ALT tended to increase, depending on the duration of treatment. Treatment with 5 and 10 mg/dl UA had an antioxidative effect on the model cells, and 30 mg/dl UA treatment for 48 h increased OS in the cells.
Collapse
Affiliation(s)
- Shi Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Yan Yang
- Department of Child Healthcare, People's Hospital (Children's Hospital) North Hospital, Urumqi, Xinjiang 830011, P.R. China
| | - Yong Zhou
- Department of Biology, School of Basic Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Wei Xiang
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Hua Yao
- Health Management Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Ling Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
119
|
Mi S, Yi X, Du Z, Xu Y, Sun W. Construction of a liver sinusoid based on the laminar flow on chip and self-assembly of endothelial cells. Biofabrication 2018; 10:025010. [DOI: 10.1088/1758-5090/aaa97e] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
120
|
Lee SH, Sung JH. Organ-on-a-Chip Technology for Reproducing Multiorgan Physiology. Adv Healthc Mater 2018; 7. [PMID: 28945001 DOI: 10.1002/adhm.201700419] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/04/2017] [Indexed: 12/14/2022]
Abstract
In the drug development process, the accurate prediction of drug efficacy and toxicity is important in order to reduce the cost, labor, and effort involved. For this purpose, conventional 2D cell culture models are used in the early phase of drug development. However, the differences between the in vitro and the in vivo systems have caused the failure of drugs in the later phase of the drug-development process. Therefore, there is a need for a novel in vitro model system that can provide accurate information for evaluating the drug efficacy and toxicity through a closer recapitulation of the in vivo system. Recently, the idea of using microtechnology for mimicking the microscale tissue environment has become widespread, leading to the development of "organ-on-a-chip." Furthermore, the system is further developed for realizing a multiorgan model for mimicking interactions between multiple organs. These advancements are still ongoing and are aimed at ultimately developing "body-on-a-chip" or "human-on-a-chip" devices for predicting the response of the whole body. This review summarizes recently developed organ-on-a-chip technologies, and their applications for reproducing multiorgan functions.
Collapse
Affiliation(s)
- Seung Hwan Lee
- School of Chemical and Biological Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering; Hongik University; Seoul 04066 Republic of Korea
| |
Collapse
|
121
|
Oseini AM, Cole BK, Issa D, Feaver RE, Sanyal AJ. Translating scientific discovery: the need for preclinical models of nonalcoholic steatohepatitis. Hepatol Int 2018; 12:6-16. [PMID: 29299759 PMCID: PMC5815925 DOI: 10.1007/s12072-017-9838-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/24/2017] [Indexed: 12/29/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the Western world, affecting about 1/3 of the US general population and remaining as a significant cause of morbidity and mortality. The hallmark of the disease is the excessive accumulation of fat within the liver cells (hepatocytes), which eventually paves the way to cellular stress, injury and apoptosis. NAFLD is strongly associated with components of the metabolic syndrome and is fast emerging as a leading cause of liver transplant in the USA. Based on clinico-pathologic classification, NAFLD may present as isolated lipid collection (steatosis) within the hepatocytes (referred to as non-alcoholic fatty liver; NAFL); or as the more aggressive phenotype (known as non-alcoholic steatohepatitis; NASH). There are currently no regulatory agency- approved medication for NAFLD, despite the enormous work and resources that have gone into the study of this condition. Therefore, there remains a huge unmet need in developing and utilizing pre-clinical models that will recapitulate the disease condition in humans. In line with progress being made in developing appropriate disease models, this review highlights the cutting-edge preclinical in vitro and animal models that try to recapitulate the human disease pathophysiology and/or clinical manifestations.
Collapse
Affiliation(s)
- Abdul M. Oseini
- Division of Gastroenterology, Department of Medicine, VCU School of Medicine, MCV Box 980341, Richmond, VA 23298-0341, USA
| | - Banumathi K. Cole
- HemoShear Therapeutics, 501 Locust Ave, Suite 301, Charlottesville, VA 22902, USA
| | - Danny Issa
- Division of Gastroenterology, Department of Medicine, VCU School of Medicine, MCV Box 980341, Richmond, VA 23298-0341, USA
| | - Ryan E. Feaver
- HemoShear Therapeutics, 501 Locust Ave, Suite 301, Charlottesville, VA 22902, USA
| | - Arun J. Sanyal
- Division of Gastroenterology, Department of Medicine, VCU School of Medicine, MCV Box 980341, Richmond, VA 23298-0341, USA
- Physiology and Molecular Pathology, MCV Box 980341, Richmond, VA 23298-0341, USA
| |
Collapse
|
122
|
Cole BK, Feaver RE, Wamhoff BR, Dash A. Non-alcoholic fatty liver disease (NAFLD) models in drug discovery. Expert Opin Drug Discov 2017; 13:193-205. [PMID: 29190166 DOI: 10.1080/17460441.2018.1410135] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The progressive disease spectrum of non-alcoholic fatty liver disease (NAFLD), which includes non-alcoholic steatohepatitis (NASH), is a rapidly emerging public health crisis with no approved therapy. The diversity of various therapies under development highlights the lack of consensus around the most effective target, underscoring the need for better translatable preclinical models to study the complex progressive disease and effective therapies. Areas covered: This article reviews published literature of various mouse models of NASH used in preclinical studies, as well as complex organotypic in vitro and ex vivo liver models being developed. It discusses translational challenges associated with both kinds of models, and describes some of the studies that validate their application in NAFLD. Expert opinion: Animal models offer advantages of understanding drug distribution and effects in a whole body context, but are limited by important species differences. Human organotypic in vitro and ex vivo models with physiological relevance and translatability need to be used in a tiered manner with simpler screens. Leveraging newer technologies, like metabolomics, proteomics, and transcriptomics, and the future development of validated disease biomarkers will allow us to fully utilize the value of these models to understand disease and evaluate novel drugs in isolation or combination.
Collapse
Affiliation(s)
| | | | | | - Ajit Dash
- b Early Development Safety , Genentech Inc , South San Francisco , CA , USA
| |
Collapse
|
123
|
McCormick S, Tong Z, Ivask A, Morozesk M, Voelcker NH, Lombi E, Priest C. Optimization of binding B-lymphocytes in a microfluidic channel: surface modification, stasis time and shear response. Biofabrication 2017; 10:014101. [DOI: 10.1088/1758-5090/aa9554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
124
|
Mandrycky C, Phong K, Zheng Y. Tissue engineering toward organ-specific regeneration and disease modeling. MRS COMMUNICATIONS 2017; 7:332-347. [PMID: 29750131 PMCID: PMC5939579 DOI: 10.1557/mrc.2017.58] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/17/2017] [Indexed: 05/17/2023]
Abstract
Tissue engineering has been recognized as a translational approach to replace damaged tissue or whole organs. Engineering tissue, however, faces an outstanding knowledge gap in the challenge to fully recapitulate complex organ-specific features. Major components, such as cells, matrix, and architecture, must each be carefully controlled to engineer tissue-specific structure and function that mimics what is found in vivo. Here we review different methods to engineer tissue, and discuss critical challenges in recapitulating the unique features and functional units in four major organs-the kidney, liver, heart, and lung, which are also the top four candidates for organ transplantation in the USA. We highlight advances in tissue engineering approaches to enable the regeneration of complex tissue and organ substitutes, and provide tissue-specific models for drug testing and disease modeling. We discuss the current challenges and future perspectives toward engineering human tissue models.
Collapse
Affiliation(s)
- Christian Mandrycky
- Departments of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kiet Phong
- Departments of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Ying Zheng
- Departments of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
125
|
Computationally Informed Design of a Multi-Axial Actuated Microfluidic Chip Device. Sci Rep 2017; 7:5489. [PMID: 28710359 PMCID: PMC5511244 DOI: 10.1038/s41598-017-05237-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/25/2017] [Indexed: 12/02/2022] Open
Abstract
This paper describes the computationally informed design and experimental validation of a microfluidic chip device with multi-axial stretching capabilities. The device, based on PDMS soft-lithography, consisted of a thin porous membrane, mounted between two fluidic compartments, and tensioned via a set of vacuum-driven actuators. A finite element analysis solver implementing a set of different nonlinear elastic and hyperelastic material models was used to drive the design and optimization of chip geometry and to investigate the resulting deformation patterns under multi-axial loading. Computational results were cross-validated by experimental testing of prototypal devices featuring the in silico optimized geometry. The proposed methodology represents a suite of computationally handy simulation tools that might find application in the design and in silico mechanical characterization of a wide range of stretchable microfluidic devices.
Collapse
|
126
|
McCormick SC, Kriel FH, Ivask A, Tong Z, Lombi E, Voelcker NH, Priest C. The Use of Microfluidics in Cytotoxicity and Nanotoxicity Experiments. MICROMACHINES 2017. [PMCID: PMC6190054 DOI: 10.3390/mi8040124] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Many unique chemical compounds and nanomaterials are being developed, and each one requires a considerable range of in vitro and/or in vivo toxicity screening in order to evaluate their safety. The current methodology of in vitro toxicological screening on cells is based on well-plate assays that require time-consuming manual handling or expensive automation to gather enough meaningful toxicology data. Cost reduction; access to faster, more comprehensive toxicity data; and a robust platform capable of quantitative testing, will be essential in evaluating the safety of new chemicals and nanomaterials, and, at the same time, in securing the confidence of regulators and end-users. Microfluidic chips offer an alternative platform for toxicity screening that has the potential to transform both the rates and efficiency of nanomaterial testing, as reviewed here. The inherent advantages of microfluidic technologies offer high-throughput screening with small volumes of analytes, parallel analyses, and low-cost fabrication.
Collapse
Affiliation(s)
- Scott C. McCormick
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
| | - Frederik H. Kriel
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
| | - Angela Ivask
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
| | - Ziqiu Tong
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052 VIC, Australia
| | - Enzo Lombi
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
| | - Nicolas H. Voelcker
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052 VIC, Australia
| | - Craig Priest
- Future Industries Institute, University of South Australia, Mawson Lakes Blvd., Mawson Lakes, 5098 SA, Australia; (S.C.M.); (F.H.K.); (A.I.); (Z.T.); (E.L.); (N.H.V.)
- Correspondence: ; Tel.: +61-8-8302-5146
| |
Collapse
|
127
|
Gu JJ, Yao M, Yang J, Cai Y, Zheng WJ, Wang L, Yao DB, Yao DF. Mitochondrial carnitine palmitoyl transferase-II inactivity aggravates lipid accumulation in rat hepatocarcinogenesis. World J Gastroenterol 2017; 23:256-264. [PMID: 28127199 PMCID: PMC5236505 DOI: 10.3748/wjg.v23.i2.256] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/29/2016] [Accepted: 10/31/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the dynamic alteration of mitochondrial carnitine palmitoyl transferase II (CPT-II) expression during malignant transformation of rat hepatocytes. METHODS Sprague-Dawley male rats were fed with normal, high fat (HF), and HF containing 2-fluorenylacetamide (2-FAA) diet, respectively. According to the Hematoxylin and Eosin staining of livers, rats were divided into control, fatty liver, degeneration, precancerous, and cancerous groups. Liver lipids were dyed with Oil Red O, CPT-II alterations were analyzed by immunohistochemistry, and compared with CPT-II specific concentration (μg/mg protein). Levels of total cholesterol (Tch), triglyceride (TG), and amino-transferases [alanine aminotransferase (ALT), aspartate aminotransferase (AST)] were determined by the routine methods. RESULTS After intake of HF and/or HF+2-FAA diets, the rat livers showed mass lipid accumulation. The lipid level in the control group was significantly lower than that in other groups. The changes of serum TG and Tch levels were abnormally increasing, 2-3 times more than those in the controls (P < 0.05). During the rat liver morphological changes from normal to cancer development process with hepatocyte injury, serum AST and ALT levels were significantly higher (4-8 times, P < 0.05) than those in the control group. The specific concentration of CPT-II in liver tissues progressively decreased during hepatocyte malignant transformation, with the lowest CPT-II levels in the cancer group than in any of the other groups (P < 0.05). CONCLUSION Low CPT-II expression might lead to abnormal hepatic lipid accumulation, which should promote the malignant transformation of hepatocytes.
Collapse
|
128
|
Feaver RE, Cole BK, Lawson MJ, Hoang SA, Marukian S, Blackman BR, Figler RA, Sanyal AJ, Wamhoff BR, Dash A. Development of an in vitro human liver system for interrogating nonalcoholic steatohepatitis. JCI Insight 2016; 1:e90954. [PMID: 27942596 DOI: 10.1172/jci.insight.90954] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A barrier to drug development for nonalcoholic steatohepatitis (NASH) is the absence of translational preclinical human-relevant systems. An in vitro liver model was engineered to incorporate hepatic sinusoidal flow, transport, and lipotoxic stress risk factors (glucose, insulin, free fatty acids) with cocultured primary human hepatocytes, hepatic stellate cells (HSCs), and macrophages. Transcriptomic, lipidomic, and functional endpoints were evaluated and compared with clinical data from NASH patient biopsies. The lipotoxic milieu promoted hepatocyte lipid accumulation (4-fold increase, P < 0.01) and a lipidomics signature similar to NASH biopsies. Hepatocyte glucose output increased with decreased insulin sensitivity. These changes were accompanied by increased inflammatory analyte secretion (e.g., IL-6, IL-8, alanine aminotransferase). Fibrogenic activation markers increased with lipotoxic conditions, including secreted TGF-β (>5-fold increase, P < 0.05), extracellular matrix gene expression, and HSC activation. Significant pathway correlation existed between this in vitro model and human biopsies. Consistent with clinical trial data, 0.5 μM obeticholic acid in this model promoted a healthy lipidomic signature, reduced inflammatory and fibrotic secreted factors, but also increased ApoB secretion, suggesting a potential adverse effect on lipoprotein metabolism. Lipotoxic stress activates similar biological signatures observed in NASH patients in this system, which may be relevant for interrogating novel therapeutic approaches to treat NASH.
Collapse
Affiliation(s)
- Ryan E Feaver
- HemoShear Therapeutics LLC, Charlottesville, Virginia, USA
| | | | - Mark J Lawson
- HemoShear Therapeutics LLC, Charlottesville, Virginia, USA
| | | | | | | | | | - Arun J Sanyal
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virgina, USA
| | | | - Ajit Dash
- HemoShear Therapeutics LLC, Charlottesville, Virginia, USA
| |
Collapse
|