401
|
Blutt SE, Broughman JR, Zou W, Zeng XL, Karandikar UC, In J, Zachos NC, Kovbasnjuk O, Donowitz M, Estes MK. Gastrointestinal microphysiological systems. Exp Biol Med (Maywood) 2017; 242:1633-1642. [PMID: 28534432 DOI: 10.1177/1535370217710638] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal diseases are a significant health care and economic burden. Prevention and treatment of these diseases have been limited by the available human biologic models. Microphysiological systems comprise organ-specific human cultures that recapitulate many structural, biological, and functional properties of the organ in smaller scale including aspects of flow, shear stress and chemical gradients. The development of intestinal microphysiological system platforms represents a critical component in improving our understanding, prevention, and treatment of gastrointestinal diseases. This minireview discusses: shortcomings of classical cell culture models of the gastrointestinal tract; human intestinal enteroids as a new model and their advantages compared to cell lines; why intestinal microphysiological systems are needed; potential functional uses of intestinal microphysiological systems in areas of drug development and modeling acute and chronic diseases; and current challenges in the development of intestinal microphysiological systems. Impact statement The development of a gastrointestinal MPS has the potential to facilitate the understanding of GI physiology. An ultimate goal is the integration of the intestinal MPS with other organ MPS. The development and characterization of nontransformed human intestinal cultures for use in MPS have progressed significantly since the inception of the MPS program in 2012, and these cultures are a key component of advancing MPS. Continued efforts are needed to optimize MPS to comprehensively and accurately recapitulate the complexity of the intestinal epithelium within intestinal tissue. These systems will need to include peristalsis, flow, and oxygen gradients, with incorporation of vascular, immune, and nerve cells. Regional cellular organization of crypt and villus areas will also be necessary to better model complete intestinal structure.
Collapse
Affiliation(s)
- Sarah E Blutt
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Broughman
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Winnie Zou
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi-Lei Zeng
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Umesh C Karandikar
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julie In
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nicholas C Zachos
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Olga Kovbasnjuk
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mark Donowitz
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mary K Estes
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.,3 Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
402
|
|
403
|
Aziz AUR, Geng C, Fu M, Yu X, Qin K, Liu B. The Role of Microfluidics for Organ on Chip Simulations. Bioengineering (Basel) 2017; 4:E39. [PMID: 28952518 PMCID: PMC5590458 DOI: 10.3390/bioengineering4020039] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022] Open
Abstract
A multichannel three-dimensional chip of a microfluidic cell culture which enables the simulation of organs is called an "organ on a chip" (OC). With the integration of many other technologies, OCs have been mimicking organs, substituting animal models, and diminishing the time and cost of experiments which is better than the preceding conventional in vitro models, which make them imperative tools for finding functional properties, pathological states, and developmental studies of organs. In this review, recent progress regarding microfluidic devices and their applications in cell cultures is discussed to explain the advantages and limitations of these systems. Microfluidics is not a solution but only an approach to create a controlled environment, however, other supporting technologies are needed, depending upon what is intended to be achieved. Microfluidic platforms can be integrated with additional technologies to enhance the organ on chip simulations. Besides, new directions and areas are mentioned for interested researchers in this field, and future challenges regarding the simulation of OCs are also discussed, which will make microfluidics more accurate and beneficial for biological applications.
Collapse
Affiliation(s)
- Aziz Ur Rehman Aziz
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Chunyang Geng
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Mengjie Fu
- Dalian Institute of Maternal and Child Health Care. Dalian 116024, Liaoning Province, China.
| | - Xiaohui Yu
- Dalian Institute of Maternal and Child Health Care. Dalian 116024, Liaoning Province, China.
| | - Kairong Qin
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Bo Liu
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| |
Collapse
|
404
|
Bae CY, Son J, Kim H, Park JK. Demonstration of Interposed Modular Hydrogel Sheet for Multicellular Analysis in a Microfluidic Assembly Platform. Sci Rep 2017; 7:1289. [PMID: 28465532 PMCID: PMC5430983 DOI: 10.1038/s41598-017-01363-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/29/2017] [Indexed: 12/29/2022] Open
Abstract
Hydrogel sheets have emerged as a promising biomaterial scaffold for the encapsulation and transfer of multicellular structures. Although the improvement of the chemical interactions and the design of micro-scaled geometry have contributed to the development of multipurpose hydrogel scaffolds, the application of hydrogel sheets to assess multicellular structures is still challenging. To expand the technical applicability of hydrogel sheets, we here demonstrate that a single layer of the hydrogel sheet can be integrated as an interposed module in a microfluidic device for multicellular analysis. As a cell culture unit, encapsulated pancreatic insulinoma (MIN6) cells in the hydrogel sheet were labeled and examined via multiple microchannels. After obtaining simultaneously multi-labeled cells in the hydrogel sheet that had been incorporated into the microfluidic device, each modular hydrogel sheet was also recoverable and re-cultured without any distortion. The modular hydrogel sheet can be simply manipulated and conserved as a multicellular module in a three-dimensional (3D) in vitro culture platform. Using the modular concept of hydrogel sheets capable of cell culture and/or assay, an integrated multicellular analysis in the microfluidic device is expected to improve accessibility, scalability, and practicality for end users.
Collapse
Affiliation(s)
- Chae Yun Bae
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jaejung Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Je-Kyun Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
405
|
Li Z, Su W, Zhu Y, Tao T, Li D, Peng X, Qin J. Drug absorption related nephrotoxicity assessment on an intestine-kidney chip. BIOMICROFLUIDICS 2017; 11:034114. [PMID: 28652884 PMCID: PMC5453794 DOI: 10.1063/1.4984768] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/18/2017] [Indexed: 05/04/2023]
Abstract
Drug absorption in the intestine is tightly related to drug-induced nephrotoxicity, which is a relatively common side effect in clinical practice. It highlights a great need to develop predictive models with high accuracy in the early stage during new drug discovery and development. Herein, we presented a novel intestine-kidney chip, which recapitulated drug absorption in the intestine and its resultant drug toxicity on the kidney. This work aims to provide an integrated tool for accurate assessment of drug absorption-related nephrotoxicity in vitro. A microfluidic device with multi-interfaces was designed, which facilitated the co-culture of the intestinal and glomerular endothelial cells in compartmentalized micro-chambers. Thus, drug absorption and following nephrotoxicity could be explored in a single assay based on the formation of the intact intestine function on the chip. Specifically, we adopt digoxin (DIG) as a model drug combined with colestyramine (COL) or Verapamil (VER), which significantly influence DIG absorption in the intestine. Different degrees of nephrotoxicity under drug combinations were further observed on the chip, including cell apoptosis, cell viability, and lactate dehydrogenase leakage. These features were consistent with the variance of DIG absorption by the intestinal cells. In agreement with clinical observations, our data demonstrated that DIG-induced nephrotoxicity was enhanced combined with VER but weakened with COL. All of these findings suggest that the established microdevice might provide a useful and cost-effective platform in vitro for testing drug absorption and nephrotoxicity in preclinical trials during new drug development.
Collapse
Affiliation(s)
| | - Wentao Su
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | | | - Tingting Tao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Dong Li
- Dalian Municipal Women and Children's Medical Center, Dalian 116024, China
| | - Xiaojun Peng
- College of Chemistry, Dalian University of Technology, Dalian 116024, China
| | | |
Collapse
|
406
|
Ribas J, Zhang YS, Pitrez PR, Leijten J, Miscuglio M, Rouwkema J, Dokmeci MR, Nissan X, Ferreira L, Khademhosseini A. Biomechanical Strain Exacerbates Inflammation on a Progeria-on-a-Chip Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:10.1002/smll.201603737. [PMID: 28211642 PMCID: PMC5545787 DOI: 10.1002/smll.201603737] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/02/2017] [Indexed: 05/22/2023]
Abstract
Organ-on-a-chip platforms seek to recapitulate the complex microenvironment of human organs using miniaturized microfluidic devices. Besides modeling healthy organs, these devices have been used to model diseases, yielding new insights into pathophysiology. Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease showing accelerated vascular aging, leading to the death of patients due to cardiovascular diseases. HGPS targets primarily vascular cells, which reside in mechanically active tissues. Here, a progeria-on-a-chip model is developed and the effects of biomechanical strain are examined in the context of vascular aging and disease. Physiological strain induces a contractile phenotype in primary smooth muscle cells (SMCs), while a pathological strain induces a hypertensive phenotype similar to that of angiotensin II treatment. Interestingly, SMCs derived from human induced pluripotent stem cells of HGPS donors (HGPS iPS-SMCs), but not from healthy donors, show an exacerbated inflammatory response to strain. In particular, increased levels of inflammation markers as well as DNA damage are observed. Pharmacological intervention reverses the strain-induced damage by shifting gene expression profile away from inflammation. The progeria-on-a-chip is a relevant platform to study biomechanics in vascular biology, particularly in the setting of vascular disease and aging, while simultaneously facilitating the discovery of new drugs and/or therapeutic targets.
Collapse
Affiliation(s)
- João Ribas
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Doctoral Program in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Patrícia R. Pitrez
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Mario Miscuglio
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Mehmet Remzi Dokmeci
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Xavier Nissan
- INSERM U861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex 91030, France
| | | | | |
Collapse
|
407
|
Lelièvre SA, Kwok T, Chittiboyina S. Architecture in 3D cell culture: An essential feature for in vitro toxicology. Toxicol In Vitro 2017; 45:287-295. [PMID: 28366709 DOI: 10.1016/j.tiv.2017.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/20/2017] [Accepted: 03/28/2017] [Indexed: 01/06/2023]
Abstract
Three-dimensional cell culture has the potential to revolutionize toxicology studies by allowing human-based reproduction of essential elements of organs. Beyond the study of toxicants on the most susceptible organs such as liver, kidney, skin, lung, gastrointestinal tract, testis, heart and brain, carcinogenesis research will also greatly benefit from 3D cell culture models representing any normal tissue. No tissue function can be suitably reproduced without the appropriate tissue architecture whether mimicking acini, ducts or tubes, sheets of cells or more complex cellular organizations like hepatic cords. In this review, we illustrate the fundamental characteristics of polarity that is an essential architectural feature of organs for which different 3D cell culture models are available for toxicology studies in vitro. The value of tissue polarity for the development of more accurate carcinogenesis studies is also exemplified, and the concept of using extracellular gradients of gaseous or chemical substances produced with microfluidics in 3D cell culture is discussed. Indeed such gradients-on-a-chip might bring unprecedented information to better determine permissible exposure levels. Finally, the impact of tissue architecture, established via cell-matrix interactions, on the cell nucleus is emphasized in light of the importance in toxicology of morphological and epigenetic alterations of this organelle.
Collapse
Affiliation(s)
- Sophie A Lelièvre
- Purdue University, Department of Basic Medical Sciences, 625 Harrison Street, West Lafayette, IN 47907, USA; 3D Cell Culture Core (3D3C) Facility, Birck Nanotechnology Center, Purdue University Discovery Park, 1205 West State Street, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, 201 S University Street, West Lafayette, IN 47907, USA.
| | - Tim Kwok
- 3D Cell Culture Core (3D3C) Facility, Birck Nanotechnology Center, Purdue University Discovery Park, 1205 West State Street, West Lafayette, IN 47907, USA
| | - Shirisha Chittiboyina
- Purdue University, Department of Basic Medical Sciences, 625 Harrison Street, West Lafayette, IN 47907, USA; 3D Cell Culture Core (3D3C) Facility, Birck Nanotechnology Center, Purdue University Discovery Park, 1205 West State Street, West Lafayette, IN 47907, USA
| |
Collapse
|
408
|
Drieschner C, Minghetti M, Wu S, Renaud P, Schirmer K. Ultrathin Alumina Membranes as Scaffold for Epithelial Cell Culture from the Intestine of Rainbow Trout. ACS APPLIED MATERIALS & INTERFACES 2017; 9:9496-9505. [PMID: 28244327 DOI: 10.1021/acsami.7b00705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Permeable membranes are indispensable for in vitro epithelial barrier models. However, currently available polymer-based membranes are low in porosity and relatively thick, resulting in a limited permeability and unrealistic culture conditions. In this study, we developed an ultrathin, nanoporous alumina membrane as novel cell culture interface for vertebrate cells, with focus on the rainbow trout (Onchorynchus mykiss) intestinal cell line RTgutGC. The new type of membrane is framed in a silicon chip for physical support and has a thickness of only 1 μm, with a porosity of 15% and homogeneous nanopores (Ø = 73 ± 21 nm). Permeability rates for small molecules, namely lucifer yellow, dextran 40, and bovine serum albumin, exceeded those of standard polyethylene terephthalate (PET) membranes by up to 27 fold. With the final goal to establish a representative model of the fish intestine for environmental toxicology, we engineered a simple culture setup, capable of testing the cellular response toward chemical exposure. Herein, cells were cultured in a monolayer on the alumina membranes and formed a polarized epithelium with apical expression of the tight junction protein ZO-1 within 14 days. Impedance spectroscopy, a noninvasive and real time electrical measurement, was used to determine cellular resistance during epithelial layer formation and chemical exposure to evaluate barrier functionality. Resistance values during epithelial development revealed different stages of epithelial maturity and were comparable with the in vivo situation. During chemical exposure, cellular resistance changed immediately when barrier tightness or cell viability was affected. Thus, our study demonstrates nanoporous alumina membranes as promising novel interface for alternative in vitro approaches, capable of allowing cell culture in a physiologically realistic manner and enabling high quality microscopy and sensitive measurement of cellular resistance.
Collapse
Affiliation(s)
- Carolin Drieschner
- Eawag, Swiss Federal Institute of Aquatic Science and Technology , 8600 Dübendorf, Switzerland
- Microsystems Laboratory 4, School of Architecture, École Polytechnique Fédérale de Lausanne , 1015 Lausanne, Switzerland
| | - Matteo Minghetti
- Eawag, Swiss Federal Institute of Aquatic Science and Technology , 8600 Dübendorf, Switzerland
- Department of Integrative Biology, Oklahoma State University , 74078 Oklahoma, United States
| | - Songmei Wu
- Microsystems Laboratory 4, School of Architecture, École Polytechnique Fédérale de Lausanne , 1015 Lausanne, Switzerland
- School of Science, Beijing Jiaotong University , 100044 Beijing, P. R China
| | - Philippe Renaud
- Microsystems Laboratory 4, School of Architecture, École Polytechnique Fédérale de Lausanne , 1015 Lausanne, Switzerland
| | - Kristin Schirmer
- Eawag, Swiss Federal Institute of Aquatic Science and Technology , 8600 Dübendorf, Switzerland
- Department of Civil and Environmental Engineering, School of Architecture, École Polytechnique Fédérale de Lausanne , 1015 Lausanne, Switzerland
- Institute of Biogeochemistry and Pollutant Dynamics, ETH-Zürich , 8093 Zürich, Switzerland
| |
Collapse
|
409
|
Lee SH, Shim KY, Kim B, Sung JH. Hydrogel-based three-dimensional cell culture for organ-on-a-chip applications. Biotechnol Prog 2017; 33:580-589. [PMID: 28247962 DOI: 10.1002/btpr.2457] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/23/2016] [Indexed: 12/29/2022]
Abstract
Recent studies have reported that three-dimensionally cultured cells have more physiologically relevant functions than two-dimensionally cultured cells. Cells are three-dimensionally surrounded by the extracellular matrix (ECM) in complex in vivo microenvironments and interact with the ECM and neighboring cells. Therefore, replicating the ECM environment is key to the successful cell culture models. Various natural and synthetic hydrogels have been used to mimic ECM environments based on their physical, chemical, and biological characteristics, such as biocompatibility, biodegradability, and biochemical functional groups. Because of these characteristics, hydrogels have been combined with microtechnologies and used in organ-on-a-chip applications to more closely recapitulate the in vivo microenvironment. Therefore, appropriate hydrogels should be selected depending on the cell types and applications. The porosity of the selected hydrogel should be controlled to facilitate the movement of nutrients and oxygen. In this review, we describe various types of hydrogels, external stimulation-based gelation of hydrogels, and control of their porosity. Then, we introduce applications of hydrogels for organ-on-a-chip. Last, we also discuss the challenges of hydrogel-based three-dimensional cell culture techniques and propose future directions. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:580-589, 2017.
Collapse
Affiliation(s)
- Seung Hwan Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 151-742, Republic of Korea
| | - Kyu Young Shim
- Chemical Engineering, Hongik University, Seoul, 121-791, Republic of Korea
| | - Bumsang Kim
- Chemical Engineering, Hongik University, Seoul, 121-791, Republic of Korea
| | - Jong Hwan Sung
- Chemical Engineering, Hongik University, Seoul, 121-791, Republic of Korea
| |
Collapse
|
410
|
Gleeson JP, Brayden DJ, Ryan SM. Evaluation of PepT1 transport of food-derived antihypertensive peptides, Ile-Pro-Pro and Leu-Lys-Pro using in vitro, ex vivo and in vivo transport models. Eur J Pharm Biopharm 2017; 115:276-284. [PMID: 28315445 DOI: 10.1016/j.ejpb.2017.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/08/2017] [Accepted: 03/11/2017] [Indexed: 11/18/2022]
Abstract
Ile-Pro-Pro (IPP) and Leu-Lys-Pro (LKP) are food-derived antihypertensive peptides which inhibit angiotensin-converting enzyme (ACE) and may have potential to attenuate hypertension. There is debate over their mechanism of uptake across small intestinal epithelia, but paracellular and PepT1 carrier-mediated uptake are thought to be important routes. The aim of this study was to determine their routes of intestinal permeability using in vitro, ex vivo and in vivo intestinal models. The presence of an apical side pH of 6.5 (mimicking the intestinal acidic microclimate) and of Gly-Sar (a high affinity competitive inhibitor and substrate for PepT1) were tested on the transepithelial apical to basolateral (A to B) transport of [3H]-IPP and [3H]-LKP across filter-grown Caco-2 monolayers in vitro and rat jejunal mucosae ex vivo. A buffer pH of 6.5 on the apical side enabled Gly-Sar to reduce the apparent permeability (Papp) of [3H]-IPP and [3H]-LKP, but this inhibition was not evident at an apical buffer pH of 7.4. Gly-Sar reduced the Papp across isolated jejunal mucosae and the area under the curve (AUC) in intra-jejunal instillations when the apical/luminal buffer pH was either 7.4 or 6.5. However, the jejunal surface acidic pH was maintained in rat jejunal tissue even when the apical side buffer pH was 7.4 due to the presence of the microclimate which is not present in monolayers. PepT1 expression was confirmed by immunofluorescence on monolayers and brush border of rat jejunal tissue. This data suggest that IPP and LKP are highly permeable and cross small intestinal epithelia in part by the PepT1 transporter, with an additional contribution from the paracellular route.
Collapse
Affiliation(s)
- John P Gleeson
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sinéad M Ryan
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
411
|
Integration concepts for multi-organ chips: how to maintain flexibility?! Future Sci OA 2017; 3:FSO180. [PMID: 28670472 PMCID: PMC5481865 DOI: 10.4155/fsoa-2016-0092] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/01/2017] [Indexed: 12/28/2022] Open
Abstract
Multi-organ platforms have an enormous potential to lead to a paradigm shift in a multitude of research domains including drug development, toxicological screening, personalized medicine as well as disease modeling. Integrating multiple organ–tissues into one microfluidic circulation merges the advantages of cell lines (human genetic background) and animal models (complex physiology) and enables the creation of more in vivo-like in vitro models. In recent years, a variety of design concepts for multi-organ platforms have been introduced, categorizable into static, semistatic and flexible systems. The most promising approach seems to be flexible interconnection of single-organ platforms to application-specific multi-organ systems. This perspective elucidates the concept of ‘mix-and-match’ toolboxes and discusses the numerous advantages compared with static/semistatic platforms as well as remaining challenges. ‘Organs-on-a-chip’ are platforms accommodating organ-specific human tissues in microscale 3D chambers with physiologically relevant structure. Broken down to the basic building blocks but simultaneously mimicking essential organ functions, these sophisticated biochips can help reduce the need for animal models in drug development, toxicity screening and basic research. However, to simulate a drug's journey through the human body, it is necessary to consider how a combination of organs responds to a given drug. In this perspective, concepts of realizing such ‘multi-organ platforms’ and the need for ‘mix-and-match’ toolboxes, which contain a range of single-organ units interconnected in individual, application-specific configurations, are discussed.
Collapse
|
412
|
A microengineered collagen scaffold for generating a polarized crypt-villus architecture of human small intestinal epithelium. Biomaterials 2017; 128:44-55. [PMID: 28288348 DOI: 10.1016/j.biomaterials.2017.03.005] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/04/2017] [Accepted: 03/04/2017] [Indexed: 02/06/2023]
Abstract
The human small intestinal epithelium possesses a distinct crypt-villus architecture and tissue polarity in which proliferative cells reside inside crypts while differentiated cells are localized to the villi. Indirect evidence has shown that the processes of differentiation and migration are driven in part by biochemical gradients of factors that specify the polarity of these cellular compartments; however, direct evidence for gradient-driven patterning of this in vivo architecture has been hampered by limitations of the in vitro systems available. Enteroid cultures are a powerful in vitro system; nevertheless, these spheroidal structures fail to replicate the architecture and lineage compartmentalization found in vivo, and are not easily subjected to gradients of growth factors. In the current work, we report the development of a micropatterned collagen scaffold with suitable extracellular matrix and stiffness to generate an in vitro self-renewing human small intestinal epithelium that replicates key features of the in vivo small intestine: a crypt-villus architecture with appropriate cell-lineage compartmentalization and an open and accessible luminal surface. Chemical gradients applied to the crypt-villus axis promoted the creation of a stem/progenitor-cell zone and supported cell migration along the crypt-villus axis. This new approach combining microengineered scaffolds, biophysical cues and chemical gradients to control the intestinal epithelium ex vivo can serve as a physiologically relevant mimic of the human small intestinal epithelium, and is broadly applicable to model other tissues that rely on gradients for physiological function.
Collapse
|
413
|
Khalid N, Kobayashi I, Nakajima M. Recent lab-on-chip developments for novel drug discovery. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 9. [DOI: 10.1002/wsbm.1381] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/11/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Nauman Khalid
- School of Food and Agricultural Sciences; University of Management and Technology; Lahore Pakistan
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences; Deakin University; Waurn Ponds Australia
- Graduate School of Life and Environmental Sciences; University of Tsukuba; Tsukuba Japan
| | - Isao Kobayashi
- Graduate School of Life and Environmental Sciences; University of Tsukuba; Tsukuba Japan
- Food Research Institute; NARO; Tsukuba Japan
| | - Mitsutoshi Nakajima
- Graduate School of Life and Environmental Sciences; University of Tsukuba; Tsukuba Japan
- Food Research Institute; NARO; Tsukuba Japan
| |
Collapse
|
414
|
Yu H, Hasan NM, In JG, Estes MK, Kovbasnjuk O, Zachos NC, Donowitz M. The Contributions of Human Mini-Intestines to the Study of Intestinal Physiology and Pathophysiology. Annu Rev Physiol 2017; 79:291-312. [PMID: 28192061 PMCID: PMC5549102 DOI: 10.1146/annurev-physiol-021115-105211] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The lack of accessibility to normal and diseased human intestine and the inability to separate the different functional compartments of the intestine even when tissue could be obtained have held back the understanding of human intestinal physiology. Clevers and his associates identified intestinal stem cells and established conditions to grow "mini-intestines" ex vivo in differentiated and undifferentiated conditions. This pioneering work has made a new model of the human intestine available and has begun making contributions to the understanding of human intestinal transport in normal physiologic conditions and the pathophysiology of intestinal diseases. However, this model is reductionist and lacks many of the complexities of normal intestine. Consequently, it is not yet possible to predict how great the advances using this model will be for understanding human physiology and pathophysiology, nor how the model will be modified to include multiple other intestinal cell types and physical forces necessary to more closely approximate normal intestine. This review describes recent studies using mini-intestines, which have readdressed previously established models of normal intestinal transport physiology and newly examined intestinal pathophysiology. The emphasis is on studies with human enteroids grown either as three-dimensional spheroids or two-dimensional monolayers. In addition, comments are provided on mouse studies in cases when human studies have not yet been described.
Collapse
Affiliation(s)
- Huimin Yu
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | - Nesrin M Hasan
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | - Julie G In
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030
| | - Olga Kovbasnjuk
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | - Nicholas C Zachos
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| |
Collapse
|
415
|
Vaessen SFC, van Lipzig MMH, Pieters RHH, Krul CAM, Wortelboer HM, van de Steeg E. Regional Expression Levels of Drug Transporters and Metabolizing Enzymes along the Pig and Human Intestinal Tract and Comparison with Caco-2 Cells. Drug Metab Dispos 2017; 45:353-360. [PMID: 28153842 DOI: 10.1124/dmd.116.072231] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/09/2017] [Indexed: 01/06/2023] Open
Abstract
Intestinal transporter proteins and metabolizing enzymes play a crucial role in the oral absorption of a wide variety of drugs. The aim of the current study was to characterize better available intestinal in vitro models by comparing expression levels of these proteins and enzymes between porcine intestine, human intestine, and Caco-2 cells. We therefore determined the absolute protein expression of 19 drug transporters and the mRNA expression of 12 metabolic enzymes along the pig intestinal tract (duodenum, jejunum, ileum; N = 4), in human intestine (jejunum; N = 9), and Caco-2 cells. Expression of the included transporters and enzymes was in general well comparable between porcine and human intestinal tissue, although breast cancer resistance protein, monocarboxylate transporter 5, multidrug resistance protein (MRP) 1, MRP1, MRP3 (∼2-fold), and organic anion-transporting polypeptide (OATP) 4A1 (∼6-fold) was higher expressed in pig compared with human jejunum. Alternatively, expression level of relevant transporter proteins (glucose transporter 1, OATP4A1, MRP2, MRP1, and OATP2B1) was significantly higher (3- to 130-fold) in Caco-2 cells compared with human jejunum. Moreover, all examined CYPs showed at least a fivefold lower gene expression in Caco-2 cells compared with human jejunum, with the smallest differences for CYP1A1 and CYP3A5 and the largest difference for CYP3A4 (871-fold higher expression in human jejunum compared with Caco-2 cells). In conclusion, a comprehensive overview is provided of the expression levels of clinically relevant transporter proteins and metabolic enzymes in porcine and human intestinal tissue and Caco-2 cells, which may assist in deciding upon the most suitable model to further improve our understanding of processes that determine intestinal absorption of compounds.
Collapse
Affiliation(s)
- Stefan F C Vaessen
- TNO, Zeist, The Netherlands (M.M.H.L, C.A.M.K, H.M.W, E.S); Institute for Risk Assessment Sciences, Utrecht, The Netherlands (R.H.H.P, J.A.S,); Research Centre Technology & Innovation; and Innovative Testing in Life sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands (S.F.C.V, R.H.H.P, C.A.M.K)
| | - Marola M H van Lipzig
- TNO, Zeist, The Netherlands (M.M.H.L, C.A.M.K, H.M.W, E.S); Institute for Risk Assessment Sciences, Utrecht, The Netherlands (R.H.H.P, J.A.S,); Research Centre Technology & Innovation; and Innovative Testing in Life sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands (S.F.C.V, R.H.H.P, C.A.M.K)
| | - Raymond H H Pieters
- TNO, Zeist, The Netherlands (M.M.H.L, C.A.M.K, H.M.W, E.S); Institute for Risk Assessment Sciences, Utrecht, The Netherlands (R.H.H.P, J.A.S,); Research Centre Technology & Innovation; and Innovative Testing in Life sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands (S.F.C.V, R.H.H.P, C.A.M.K)
| | - Cyrille A M Krul
- TNO, Zeist, The Netherlands (M.M.H.L, C.A.M.K, H.M.W, E.S); Institute for Risk Assessment Sciences, Utrecht, The Netherlands (R.H.H.P, J.A.S,); Research Centre Technology & Innovation; and Innovative Testing in Life sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands (S.F.C.V, R.H.H.P, C.A.M.K)
| | - Heleen M Wortelboer
- TNO, Zeist, The Netherlands (M.M.H.L, C.A.M.K, H.M.W, E.S); Institute for Risk Assessment Sciences, Utrecht, The Netherlands (R.H.H.P, J.A.S,); Research Centre Technology & Innovation; and Innovative Testing in Life sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands (S.F.C.V, R.H.H.P, C.A.M.K)
| | - Evita van de Steeg
- TNO, Zeist, The Netherlands (M.M.H.L, C.A.M.K, H.M.W, E.S); Institute for Risk Assessment Sciences, Utrecht, The Netherlands (R.H.H.P, J.A.S,); Research Centre Technology & Innovation; and Innovative Testing in Life sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands (S.F.C.V, R.H.H.P, C.A.M.K)
| |
Collapse
|
416
|
|
417
|
Organs-on-chips: research and commercial perspectives. Drug Discov Today 2017; 22:397-403. [DOI: 10.1016/j.drudis.2016.11.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 09/30/2016] [Accepted: 11/07/2016] [Indexed: 11/19/2022]
|
418
|
Villenave R, Wales SQ, Hamkins-Indik T, Papafragkou E, Weaver JC, Ferrante TC, Bahinski A, Elkins CA, Kulka M, Ingber DE. Human Gut-On-A-Chip Supports Polarized Infection of Coxsackie B1 Virus In Vitro. PLoS One 2017; 12:e0169412. [PMID: 28146569 PMCID: PMC5287454 DOI: 10.1371/journal.pone.0169412] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/17/2016] [Indexed: 12/29/2022] Open
Abstract
Analysis of enterovirus infection is difficult in animals because they express different virus receptors than humans, and static cell culture systems do not reproduce the physical complexity of the human intestinal epithelium. Here, using coxsackievirus B1 (CVB1) as a prototype enterovirus strain, we demonstrate that human enterovirus infection, replication and infectious virus production can be analyzed in vitro in a human Gut-on-a-Chip microfluidic device that supports culture of highly differentiated human villus intestinal epithelium under conditions of fluid flow and peristalsis-like motions. When CVB1 was introduced into the epithelium-lined intestinal lumen of the device, virions entered the epithelium, replicated inside the cells producing detectable cytopathic effects (CPEs), and both infectious virions and inflammatory cytokines were released in a polarized manner from the cell apex, as they could be detected in the effluent from the epithelial microchannel. When the virus was introduced via a basal route of infection (by inoculating virus into fluid flowing through a parallel lower 'vascular' channel separated from the epithelial channel by a porous membrane), significantly lower viral titers, decreased CPEs, and delayed caspase-3 activation were observed; however, cytokines continued to be secreted apically. The presence of continuous fluid flow through the epithelial lumen also resulted in production of a gradient of CPEs consistent with the flow direction. Thus, the human Gut-on-a-Chip may provide a suitable in vitro model for enteric virus infection and for investigating mechanisms of enterovirus pathogenesis.
Collapse
Affiliation(s)
- Remi Villenave
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
| | - Samantha Q. Wales
- Molecular Virology Team, Division of Molecular Biology, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| | - Tiama Hamkins-Indik
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
| | - Efstathia Papafragkou
- Molecular Virology Team, Division of Molecular Biology, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| | - James C. Weaver
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
| | - Thomas C. Ferrante
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
| | - Christopher A. Elkins
- Molecular Virology Team, Division of Molecular Biology, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| | - Michael Kulka
- Molecular Virology Team, Division of Molecular Biology, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts, United States of America
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
419
|
Orbach SM, Less RR, Kothari A, Rajagopalan P. In Vitro Intestinal and Liver Models for Toxicity Testing. ACS Biomater Sci Eng 2017; 3:1898-1910. [PMID: 33440548 DOI: 10.1021/acsbiomaterials.6b00699] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The human body is exposed to hundreds of chemicals every day. Many of these toxicants have unknown effects on the body that can be deleterious. Furthermore, chemicals can have a synergistic effect, resulting in toxic responses of cocktails at relatively low individual exposure levels. The gastrointestinal (GI) tract and the liver are the first organs to be exposed to ingested pharmaceuticals and environmental chemicals. As a result, these organs often experience extensive damage from xenobiotics and their metabolites. In vitro models offer a promising method for testing toxic effects. Many advanced in vitro models have been developed for GI and liver toxicity. These models strive to recapitulate the in vivo organ architecture to more accurately model chemical toxicity. In this review, we discuss many of these advances, in addition to recent efforts to integrate the GI and the liver in vitro for a more holistic toxicity model.
Collapse
Affiliation(s)
- Sophia M Orbach
- Department of Chemical Engineering, ‡School of Biomedical Engineering and Sciences, and §ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Rebekah R Less
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, and §ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Anjaney Kothari
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, and ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, and ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
420
|
Compartmentalized Culture of Perivascular Stroma and Endothelial Cells in a Microfluidic Model of the Human Endometrium. Ann Biomed Eng 2017; 45:1758-1769. [PMID: 28108942 PMCID: PMC5489603 DOI: 10.1007/s10439-017-1797-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022]
Abstract
The endometrium is the inner lining of the uterus. Following specific cyclic hormonal stimulation, endometrial stromal fibroblasts (stroma) and vascular endothelial cells exhibit morphological and biochemical changes to support embryo implantation and regulate vascular function, respectively. Herein, we integrated a resin-based porous membrane in a dual chamber microfluidic device in polydimethylsiloxane that allows long term in vitro co-culture of human endometrial stromal and endothelial cells. This transparent, 2-μm porous membrane separates the two chambers, allows for the diffusion of small molecules and enables high resolution bright field and fluorescent imaging. Within our primary human co-culture model of stromal and endothelial cells, we simulated the temporal hormone changes occurring during an idealized 28-day menstrual cycle. We observed the successful differentiation of stroma into functional decidual cells, determined by morphology as well as biochemically as measured by increased production of prolactin. By controlling the microfluidic properties of the device, we additionally found that shear stress forces promoted cytoskeleton alignment and tight junction formation in the endothelial layer. Finally, we demonstrated that the endometrial perivascular stroma model was sustainable for up to 4 weeks, remained sensitive to steroids and is suitable for quantitative biochemical analysis. Future utilization of this device will allow the direct evaluation of paracrine and endocrine crosstalk between these two cell types as well as studies of immunological events associated with normal vs. disease-related endometrial microenvironments.
Collapse
|
421
|
Chang SY, Voellinger JL, Van Ness KP, Chapron B, Shaffer RM, Neumann T, White CC, Kavanagh TJ, Kelly EJ, Eaton DL. Characterization of rat or human hepatocytes cultured in microphysiological systems (MPS) to identify hepatotoxicity. Toxicol In Vitro 2017; 40:170-183. [PMID: 28089783 DOI: 10.1016/j.tiv.2017.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/05/2017] [Accepted: 01/11/2017] [Indexed: 12/12/2022]
Abstract
The liver is the main site for drug and xenobiotics metabolism, including inactivation or bioactivation. In order to improve the predictability of drug safety and efficacy in clinical development, and to facilitate the evaluation of the potential human health effects from exposure to environmental contaminants, there is a critical need to accurately model human organ systems such as the liver in vitro. We are developing a microphysiological system (MPS) based on a new commercial microfluidic platform (Nortis, Inc.) that can utilize primary liver cells from multiple species (e.g., rat and human). Compared to conventional monolayer cell culture, which typically survives for 5-7days or less, primary rat or human hepatocytes in an MPS exhibited higher viability and improved hepatic functions, such as albumin production, expression of hepatocyte marker HNF4α and canaliculi structure, for up to 14days. Additionally, induction of Cytochrome P450 (CYP) 1A and 3A4 in cryopreserved human hepatocytes was observed in the MPS. The acute cytotoxicity of the potent hepatotoxic and hepatocarcinogen, aflatoxin B1, was evaluated in human hepatocytes cultured in an MPS, demonstrating the utility of this model for acute hepatotoxicity assessment. These results indicate that MPS-cultured hepatocytes provide a promising approach for evaluating chemical toxicity in vitro.
Collapse
Affiliation(s)
- Shih-Yu Chang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jenna L Voellinger
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Kirk P Van Ness
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Brian Chapron
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Rachel M Shaffer
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | | | - Collin C White
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - David L Eaton
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
422
|
Borovjagin AV, Ogle BM, Berry JL, Zhang J. From Microscale Devices to 3D Printing: Advances in Fabrication of 3D Cardiovascular Tissues. Circ Res 2017; 120:150-165. [PMID: 28057791 PMCID: PMC5224928 DOI: 10.1161/circresaha.116.308538] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/03/2016] [Accepted: 10/19/2016] [Indexed: 01/14/2023]
Abstract
Current strategies for engineering cardiovascular cells and tissues have yielded a variety of sophisticated tools for studying disease mechanisms, for development of drug therapies, and for fabrication of tissue equivalents that may have application in future clinical use. These efforts are motivated by the need to extend traditional 2-dimensional (2D) cell culture systems into 3D to more accurately replicate in vivo cell and tissue function of cardiovascular structures. Developments in microscale devices and bioprinted 3D tissues are beginning to supplant traditional 2D cell cultures and preclinical animal studies that have historically been the standard for drug and tissue development. These new approaches lend themselves to patient-specific diagnostics, therapeutics, and tissue regeneration. The emergence of these technologies also carries technical challenges to be met before traditional cell culture and animal testing become obsolete. Successful development and validation of 3D human tissue constructs will provide powerful new paradigms for more cost effective and timely translation of cardiovascular tissue equivalents.
Collapse
Affiliation(s)
- Anton V Borovjagin
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham (A.V.B., J.L.B., J.Z.); and Department of Biomedical Engineering, College of Science and Engineering, The University of Minnesota, Minneapolis (B.M.O.)
| | - Brenda M Ogle
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham (A.V.B., J.L.B., J.Z.); and Department of Biomedical Engineering, College of Science and Engineering, The University of Minnesota, Minneapolis (B.M.O.)
| | - Joel L Berry
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham (A.V.B., J.L.B., J.Z.); and Department of Biomedical Engineering, College of Science and Engineering, The University of Minnesota, Minneapolis (B.M.O.)
| | - Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham (A.V.B., J.L.B., J.Z.); and Department of Biomedical Engineering, College of Science and Engineering, The University of Minnesota, Minneapolis (B.M.O.).
| |
Collapse
|
423
|
|
424
|
Three-Dimensional Tissue Models and Available Probes for Multi-Parametric Live Cell Microscopy: A Brief Overview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1035:49-67. [DOI: 10.1007/978-3-319-67358-5_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
425
|
Horayama M, Shinha K, Kabayama K, Fujii T, Kimura H. Spatial Chemical Stimulation Control in Microenvironment by Microfluidic Probe Integrated Device for Cell-Based Assay. PLoS One 2016; 11:e0168158. [PMID: 27930750 PMCID: PMC5145238 DOI: 10.1371/journal.pone.0168158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/24/2016] [Indexed: 12/15/2022] Open
Abstract
Cell—cell interactions play an important role in the development and function of multicellular organisms. To investigate these interactions in detail, it is necessary to evaluate the behavior of a cell population when the minimum number of cells in the population is stimulated by some chemical factors. We propose a microfluidic device integrated with microfluidic probe (MFP) functionality; this device is capable of imparting a chemical stimulus to cells within a microenvironment, for cell-based assays. The device contains MFP channels at the walls of the cell culture microchannels, and it can control a localized chemical stimulation area at the scale of a single cell to a few cells using MFP fluid control in a microspace. The results of a finite element method-based simulation indicated that it is possible to control the chemical stimulation area at the scale of a single cell to a few cells by optimizing the MFP channel apex width and the flow ratio. In addition, localized cell staining was demonstrated successfully using a spatial chemical stimulus. We confirmed the device functionality as a novel cell-based assay tool. We succeeded in performing localized cell collection using this method, which suggested that the single cell analysis of a cell monolayer that is subjected to a specific chemical stimulus is possible. The method proposed in this paper can contribute significantly to the fields of cell biology and drug development.
Collapse
Affiliation(s)
- Masayuki Horayama
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Kenta Shinha
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Osaka University, Toyonaka, Osaka, Japan
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Teruo Fujii
- Institute of Industrial Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - Hiroshi Kimura
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Kanagawa, Japan
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
426
|
Lundquist P, Artursson P. Oral absorption of peptides and nanoparticles across the human intestine: Opportunities, limitations and studies in human tissues. Adv Drug Deliv Rev 2016; 106:256-276. [PMID: 27496705 DOI: 10.1016/j.addr.2016.07.007] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/02/2016] [Accepted: 07/08/2016] [Indexed: 12/23/2022]
Abstract
In this contribution, we review the molecular and physiological barriers to oral delivery of peptides and nanoparticles. We discuss the opportunities and predictivity of various in vitro systems with special emphasis on human intestine in Ussing chambers. First, the molecular constraints to peptide absorption are discussed. Then the physiological barriers to peptide delivery are examined. These include the gastric and intestinal environment, the mucus barrier, tight junctions between epithelial cells, the enterocytes of the intestinal epithelium, and the subepithelial tissue. Recent data from human proteome studies are used to provide information about the protein expression profiles of the different physiological barriers to peptide and nanoparticle absorption. Strategies that have been employed to increase peptide absorption across each of the barriers are discussed. Special consideration is given to attempts at utilizing endogenous transcytotic pathways. To reliably translate in vitro data on peptide or nanoparticle permeability to the in vivo situation in a human subject, the in vitro experimental system needs to realistically capture the central aspects of the mentioned barriers. Therefore, characteristics of common in vitro cell culture systems are discussed and compared to those of human intestinal tissues. Attempts to use the cell and tissue models for in vitro-in vivo extrapolation are reviewed.
Collapse
Affiliation(s)
- P Lundquist
- Department of Pharmacy, Uppsala University, Box 580, SE-752 37 Uppsala, Sweden.
| | - P Artursson
- Department of Pharmacy, Uppsala University, Box 580, SE-752 37 Uppsala, Sweden.
| |
Collapse
|
427
|
Dawson A, Dyer C, Macfie J, Davies J, Karsai L, Greenman J, Jacobsen M. A microfluidic chip based model for the study of full thickness human intestinal tissue using dual flow. BIOMICROFLUIDICS 2016; 10:064101. [PMID: 27822333 PMCID: PMC5097047 DOI: 10.1063/1.4964813] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/30/2016] [Indexed: 05/09/2023]
Abstract
The study of inflammatory bowel disease, including Ulcerative Colitis and Crohn's Disease, has relied largely upon the use of animal or cell culture models; neither of which can represent all aspects of the human pathophysiology. Presented herein is a dual flow microfluidic device which holds full thickness human intestinal tissue in a known orientation. The luminal and serosal sides are independently perfused ex vivo with nutrients with simultaneous waste removal for up to 72 h. The microfluidic device maintains the viability and integrity of the tissue as demonstrated through Haematoxylin & Eosin staining, immunohistochemistry and release of lactate dehydrogenase. In addition, the inflammatory state remains in the tissue after perfusion on the device as determined by measuring calprotectin levels. It is anticipated that this human model will be extremely useful for studying the biology and testing novel interventions in diseased tissue.
Collapse
Affiliation(s)
- A Dawson
- Faculty of Life Sciences, University of Hull , Cottingham Road, Hull HU6 7RX, United Kingdom
| | - C Dyer
- Faculty of Life Sciences, University of Hull , Cottingham Road, Hull HU6 7RX, United Kingdom
| | - J Macfie
- Scarborough Hospital , Woodlands Drive, Scarborough Y012 6QL, United Kingdom
| | - J Davies
- General Surgery, Castlehill Hospital , Castle Rd, Cottingham HU16 5JQ, United Kingdom
| | - L Karsai
- Pathology Building, Hull Royal Infirmary , Anlaby Road, Hull HU3 2JZ, United Kingdom
| | - J Greenman
- Faculty of Life Sciences, University of Hull , Cottingham Road, Hull HU6 7RX, United Kingdom
| | | |
Collapse
|
428
|
|
429
|
Pereira JFS, Awatade NT, Loureiro CA, Matos P, Amaral MD, Jordan P. The third dimension: new developments in cell culture models for colorectal research. Cell Mol Life Sci 2016; 73:3971-89. [PMID: 27147463 PMCID: PMC11108567 DOI: 10.1007/s00018-016-2258-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/20/2016] [Accepted: 04/28/2016] [Indexed: 12/23/2022]
Abstract
Cellular models are important tools in various research areas related to colorectal biology and associated diseases. Herein, we review the most widely used cell lines and the different techniques to grow them, either as cell monolayer, polarized two-dimensional epithelia on membrane filters, or as three-dimensional spheres in scaffold-free or matrix-supported culture conditions. Moreover, recent developments, such as gut-on-chip devices or the ex vivo growth of biopsy-derived organoids, are also discussed. We provide an overview on the potential applications but also on the limitations for each of these techniques, while evaluating their contribution to provide more reliable cellular models for research, diagnostic testing, or pharmacological validation related to colon physiology and pathophysiology.
Collapse
Affiliation(s)
- Joana F S Pereira
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Nikhil T Awatade
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Cláudia A Loureiro
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Paulo Matos
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Margarida D Amaral
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Peter Jordan
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016, Lisbon, Portugal.
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
430
|
|
431
|
Choi JH, Lee J, Shin W, Choi JW, Kim HJ. Priming nanoparticle-guided diagnostics and therapeutics towards human organs-on-chips microphysiological system. NANO CONVERGENCE 2016; 3:24. [PMID: 28191434 PMCID: PMC5271165 DOI: 10.1186/s40580-016-0084-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/13/2016] [Indexed: 05/17/2023]
Abstract
Nanotechnology and bioengineering have converged over the past decades, by which the application of multi-functional nanoparticles (NPs) has been emerged in clinical and biomedical fields. The NPs primed to detect disease-specific biomarkers or to deliver biopharmaceutical compounds have beena validated in conventional in vitro culture models including two dimensional (2D) cell cultures or 3D organoid models. However, a lack of experimental models that have strong human physiological relevance has hampered accurate validation of the safety and functionality of NPs. Alternatively, biomimetic human "Organs-on-Chips" microphysiological systems have recapitulated the mechanically dynamic 3D tissue interface of human organ microenvironment, in which the transport, cytotoxicity, biocompatibility, and therapeutic efficacy of NPs and their conjugates may be more accurately validated. Finally, integration of NP-guided diagnostic detection and targeted nanotherapeutics in conjunction with human organs-on-chips can provide a novel avenue to accelerate the NP-based drug development process as well as the rapid detection of cellular secretomes associated with pathophysiological processes.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
| | - Jaewon Lee
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, Seoul, 04107 Republic of Korea
- Interdisciplinary Program of Integrated Biotechnology, Sogang University, Seoul, 04107 Republic of Korea
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712 USA
- School of Medicine, Pusan National University, Yangsan, 50612 Republic of Korea
| |
Collapse
|
432
|
Effect of flow and peristaltic mixing on bacterial growth in a gut-like channel. Proc Natl Acad Sci U S A 2016; 113:11414-11419. [PMID: 27681630 DOI: 10.1073/pnas.1601306113] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ecology of microbes in the gut has been shown to play important roles in the health of the host. To better understand microbial growth and population dynamics in the proximal colon, the primary region of bacterial growth in the gut, we built and applied a fluidic channel that we call the "minigut." This is a channel with an array of membrane valves along its length, which allows mimicking active contractions of the colonic wall. Repeated contraction is shown to be crucial in maintaining a steady-state bacterial population in the device despite strong flow along the channel that would otherwise cause bacterial washout. Depending on the flow rate and the frequency of contractions, the bacterial density profile exhibits varying spatial dependencies. For a synthetic cross-feeding community, the species abundance ratio is also strongly affected by mixing and flow along the length of the device. Complex mixing dynamics due to contractions is described well by an effective diffusion term. Bacterial dynamics is captured by a simple reaction-diffusion model without adjustable parameters. Our results suggest that flow and mixing play a major role in shaping the microbiota of the colon.
Collapse
|
433
|
Pistollato F, Sumalla Cano S, Elio I, Masias Vergara M, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev 2016; 74:624-34. [DOI: 10.1093/nutrit/nuw023] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
434
|
Picollet-D’hahan N, Dolega ME, Liguori L, Marquette C, Le Gac S, Gidrol X, Martin DK. A 3D Toolbox to Enhance Physiological Relevance of Human Tissue Models. Trends Biotechnol 2016; 34:757-769. [DOI: 10.1016/j.tibtech.2016.06.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/17/2016] [Accepted: 06/28/2016] [Indexed: 01/21/2023]
|
435
|
Pensabene V, Costa L, Terekhov A, Gnecco JS, Wikswo J, Hofmeister W. Ultrathin Polymer Membranes with Patterned, Micrometric Pores for Organs-on-Chips. ACS APPLIED MATERIALS & INTERFACES 2016; 8:22629-36. [PMID: 27513606 PMCID: PMC5131702 DOI: 10.1021/acsami.6b05754] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The basal lamina or basement membrane (BM) is a key physiological system that participates in physicochemical signaling between tissue types. Its formation and function are essential in tissue maintenance, growth, angiogenesis, disease progression, and immunology. In vitro models of the BM (e.g., Boyden and transwell chambers) are common in cell biology and lab-on-a-chip devices where cells require apical and basolateral polarization. Extravasation, intravasation, membrane transport of chemokines, cytokines, chemotaxis of cells, and other key functions are routinely studied in these models. The goal of the present study was to integrate a semipermeable ultrathin polymer membrane with precisely positioned pores of 2 μm diameter in a microfluidic device with apical and basolateral chambers. We selected poly(l-lactic acid) (PLLA), a transparent biocompatible polymer, to prepare the semipermeable ultrathin membranes. The pores were generated by pattern transfer using a three-step method coupling femtosecond laser machining, polymer replication, and spin coating. Each step of the fabrication process was characterized by scanning electron microscopy to investigate reliability of the process and fidelity of pattern transfer. In order to evaluate the compatibility of the fabrication method with organs-on-a-chip technology, porous PLLA membranes were embedded in polydimethylsiloxane (PDMS) microfluidic devices and used to grow human umbilical vein endothelial cells (HUVECS) on top of the membrane with perfusion through the basolateral chamber. Viability of cells, optical transparency of membranes and strong adhesion of PLLA to PDMS were observed, thus confirming the suitability of the prepared membranes for use in organs-on-a-chip devices.
Collapse
Affiliation(s)
- Virginia Pensabene
- Department of Biomedical Engineering; Vanderbilt University, Nashville, TN 37235 USA
- School of Electronic and Electrical Engineering, University of Leeds, Leeds, LS2 9JT, UK
- School of Medicine, Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, LS9 7TF, UK
| | - Lino Costa
- Center for Laser Applications, University of Tennessee Space Institute, Tullahoma, TN 37388 USA
| | - Alexander Terekhov
- Center for Laser Applications, University of Tennessee Space Institute, Tullahoma, TN 37388 USA
| | - Juan S. Gnecco
- Department of Cellular and Molecular Pathology, Vanderbilt University, Nashville, TN 37235 USA
| | - John Wikswo
- Department of Biomedical Engineering; Vanderbilt University, Nashville, TN 37235 USA
- Vanderbilt Institute for Integrative Biosystems Research and Education; Vanderbilt University, Nashville, TN 37235 USA
- Department of Physics and Astronomy; Vanderbilt University, Nashville, TN 37235 USA
- Department of Molecular Physiology and Biophysics; Vanderbilt University, Nashville, TN 37235 USA
| | - William Hofmeister
- Center for Laser Applications, University of Tennessee Space Institute, Tullahoma, TN 37388 USA
- Vanderbilt Institute for Integrative Biosystems Research and Education; Vanderbilt University, Nashville, TN 37235 USA
| |
Collapse
|
436
|
Kim HJ, Lee J, Choi JH, Bahinski A, Ingber DE. Co-culture of Living Microbiome with Microengineered Human Intestinal Villi in a Gut-on-a-Chip Microfluidic Device. J Vis Exp 2016. [PMID: 27684630 DOI: 10.3791/54344] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Here, we describe a protocol to perform long-term co-culture of multi-species human gut microbiome with microengineered intestinal villi in a human gut-on-a-chip microphysiological device. We recapitulate the intestinal lumen-capillary tissue interface in a microfluidic device, where physiological mechanical deformations and fluid shear flow are constantly applied to mimic peristalsis. In the lumen microchannel, human intestinal epithelial Caco-2 cells are cultured to form a 'germ-free' villus epithelium and regenerate small intestinal villi. Pre-cultured microbial cells are inoculated into the lumen side to establish a host-microbe ecosystem. After microbial cells adhere to the apical surface of the villi, fluid flow and mechanical deformations are resumed to produce a steady-state microenvironment in which fresh culture medium is constantly supplied and unbound bacteria (as well as bacterial wastes) are continuously removed. After extended co-culture from days to weeks, multiple microcolonies are found to be randomly located between the villi, and both microbial and epithelial cells remain viable and functional for at least one week in culture. Our co-culture protocol can be adapted to provide a versatile platform for other host-microbiome ecosystems that can be found in various human organs, which may facilitate in vitro study of the role of human microbiome in orchestrating health and disease.
Collapse
Affiliation(s)
- Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin;
| | - Jaewon Lee
- Department of Biomedical Engineering, The University of Texas at Austin
| | - Jin-Ha Choi
- Department of Biomedical Engineering, The University of Texas at Austin
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering at Harvard University
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School; John A. Paulson School of Engineering and Applied Sciences, Harvard University
| |
Collapse
|
437
|
The motility of Entamoeba histolytica: finding ways to understand intestinal amoebiasis. Curr Opin Microbiol 2016; 34:24-30. [PMID: 27497052 DOI: 10.1016/j.mib.2016.07.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 12/16/2022]
Abstract
The pathogenic amoeba Entamoeba histolytica is able to migrate within various compartments of the human body. The present article reviews progress in understanding the mechanisms of cell motility in E. histolytica during human intestinal invasion and, in particular, how the three-dimensional characteristics of the environment regulate the parasite's behaviour. The amoeboid mode of migration that applies to E. histolytica's displacements on two-dimensional surfaces is also expected to apply to the three-dimensional environment in the human intestine although several unknown, distinct modalities may be involved. Recent advances in the field of tissue engineering have provided clues on how the construction of a human colon model could help us to understand the host's intestinal physiology and its changes following amoebic infection.
Collapse
|
438
|
Shatkin JA, Ong KJ. Alternative Testing Strategies for Nanomaterials: State of the Science and Considerations for Risk Analysis. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2016; 36:1564-1580. [PMID: 27273523 DOI: 10.1111/risa.12642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 03/02/2016] [Accepted: 04/19/2016] [Indexed: 06/06/2023]
Abstract
The rapid growth of the nanotechnology industry has warranted equal progress in the nanotoxicology and risk assessment fields. In vivo models have traditionally been used to determine human and environmental risk for chemicals; however, the use of these tests has limitations, and there are global appeals to develop reliable alternatives to animal testing. Many have investigated the use of alternative (nonanimal) testing methods and strategies have quickly developed and resulted in the generation of large toxicological data sets for numerous nanomaterials (NMs). Due to the novel physicochemical properties of NMs that are related to surface characteristics, the approach toward toxicity test development has distinct considerations from traditional chemicals, bringing new requirements for adapting these approaches for NMs. The methodical development of strategies that combine multiple alternative tests can be useful for predictive NM risk assessment and help screening-level decision making. This article provides an overview of the main developments in alternative methods and strategies for reducing uncertainty in NM risk assessment, including advantages and disadvantages of in vitro, ex vivo, and in silico methods, and examples of existing comprehensive strategies. In addition, knowledge gaps are identified toward improvements for experimental and strategy design, specifically highlighting the need to represent realistic exposure scenarios and to consider NM-specific concerns such as characterization, assay interferences, and standardization. Overall, this article aims to improve the reliability and utility of alternative testing methods and strategies for risk assessment of manufactured NMs.
Collapse
Affiliation(s)
| | - K J Ong
- Vireo Advisors, LLC, Boston, MA, USA
| |
Collapse
|
439
|
Lee J, Choi JH, Kim HJ. Human gut-on-a-chip technology: will this revolutionize our understanding of IBD and future treatments? Expert Rev Gastroenterol Hepatol 2016; 10:883-5. [PMID: 27291426 DOI: 10.1080/17474124.2016.1200466] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jaewon Lee
- a Department of Biomedical Engineering , The University of Texas at Austin , Austin , TX , USA
| | - Jin-Ha Choi
- a Department of Biomedical Engineering , The University of Texas at Austin , Austin , TX , USA
| | - Hyun Jung Kim
- a Department of Biomedical Engineering , The University of Texas at Austin , Austin , TX , USA
| |
Collapse
|
440
|
Emerging Technologies for Gut Microbiome Research. Trends Microbiol 2016; 24:887-901. [PMID: 27426971 DOI: 10.1016/j.tim.2016.06.008] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/06/2016] [Accepted: 06/23/2016] [Indexed: 02/06/2023]
Abstract
Understanding the importance of the gut microbiome on modulation of host health has become a subject of great interest for researchers across disciplines. As an intrinsically multidisciplinary field, microbiome research has been able to reap the benefits of technological advancements in systems and synthetic biology, biomaterials engineering, and traditional microbiology. Gut microbiome research has been revolutionized by high-throughput sequencing technology, permitting compositional and functional analyses that were previously an unrealistic undertaking. Emerging technologies, including engineered organoids derived from human stem cells, high-throughput culturing, and microfluidics assays allowing for the introduction of novel approaches, will improve the efficiency and quality of microbiome research. Here, we discuss emerging technologies and their potential impact on gut microbiome studies.
Collapse
|
441
|
Organ-on-chip models: new opportunities for biomedical research. Future Sci OA 2016; 3:FSO130. [PMID: 28670461 PMCID: PMC5481808 DOI: 10.4155/fsoa-2016-0038] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/10/2023] Open
|
442
|
Kang TH, Kim HJ. Farewell to Animal Testing: Innovations on Human Intestinal Microphysiological Systems. MICROMACHINES 2016; 7:mi7070107. [PMID: 30404281 PMCID: PMC6190004 DOI: 10.3390/mi7070107] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/21/2016] [Accepted: 06/21/2016] [Indexed: 02/06/2023]
Abstract
The human intestine is a dynamic organ where the complex host-microbe interactions that orchestrate intestinal homeostasis occur. Major contributing factors associated with intestinal health and diseases include metabolically-active gut microbiota, intestinal epithelium, immune components, and rhythmical bowel movement known as peristalsis. Human intestinal disease models have been developed; however, a considerable number of existing models often fail to reproducibly predict human intestinal pathophysiology in response to biological and chemical perturbations or clinical interventions. Intestinal organoid models have provided promising cytodifferentiation and regeneration, but the lack of luminal flow and physical bowel movements seriously hamper mimicking complex host-microbe crosstalk. Here, we discuss recent advances of human intestinal microphysiological systems, such as the biomimetic human "Gut-on-a-Chip" that can employ key intestinal components, such as villus epithelium, gut microbiota, and immune components under peristalsis-like motions and flow, to reconstitute the transmural 3D lumen-capillary tissue interface. By encompassing cutting-edge tools in microfluidics, tissue engineering, and clinical microbiology, gut-on-a-chip has been leveraged not only to recapitulate organ-level intestinal functions, but also emulate the pathophysiology of intestinal disorders, such as chronic inflammation. Finally, we provide potential perspectives of the next generation microphysiological systems as a personalized platform to validate the efficacy, safety, metabolism, and therapeutic responses of new drug compounds in the preclinical stage.
Collapse
Affiliation(s)
- Tae Hyun Kang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
443
|
Fröhlich E, Roblegg E. Oral uptake of nanoparticles: human relevance and the role of in vitro systems. Arch Toxicol 2016; 90:2297-314. [PMID: 27342244 DOI: 10.1007/s00204-016-1765-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 06/14/2016] [Indexed: 01/03/2023]
Abstract
Nanoparticles (NPs) present in environment, consumer and health products, food and medical applications lead to a high degree of human exposure and concerns about potential adverse effects on human health. For the general population, the exposure through contact with the skin, inhalation and oral uptake are most relevant. Since in vivo testing is only partly able to study the effects of human oral exposure, physiologically relevant in vitro systems are being developed. This review compared the three routes taking into account the estimated concentration, size of the exposed area, morphology of the involved barrier and translocation rate. The high amounts of NPs in food, the large absorption area and the relatively high translocation rate identified oral uptake as most important portal of entry for NPs into the body. Changes of NP properties in the physiological fluids, mechanisms to cross mucus and epithelial barrier, and important issues in the use of laboratory animals for oral exposure are mentioned. The ability of in vitro models to address the varying conditions along the oro-gastrointestinal tract is discussed, and requirements for physiologically relevant in vitro testing of orally ingested NPs are listed.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Stiftingtalstr. 24, 8010, Graz, Austria.
| | - Eva Roblegg
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology, Karl-Franzens-University of Graz, Graz, Austria
| |
Collapse
|
444
|
Date AA, Hanes J, Ensign LM. Nanoparticles for oral delivery: Design, evaluation and state-of-the-art. J Control Release 2016; 240:504-526. [PMID: 27292178 DOI: 10.1016/j.jconrel.2016.06.016] [Citation(s) in RCA: 287] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023]
Abstract
The oral route is a preferred method of drug administration, though achieving effective drug delivery and minimizing off-target side effects is often challenging. Formulation into nanoparticles can improve drug stability in the harsh gastrointestinal (GI) tract environment, providing opportunities for targeting specific sites in the GI tract, increasing drug solubility and bioavailability, and providing sustained release in the GI tract. However, the unique and diverse physiology throughout the GI tract, including wide variation in pH, mucus that varies in thickness and structure, numerous cell types, and various physiological functions are both a barrier to effective delivery and an opportunity for nanoparticle design. Here, nanoparticle design aspects to improve delivery to particular sites in the GI tract are discussed. We then review new methods for evaluating oral nanoparticle formulations, including a short commentary on data interpretation and translation. Finally, the state-of-the-art in preclinical targeted nanoparticle design is reviewed.
Collapse
Affiliation(s)
- Abhijit A Date
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
| | - Justin Hanes
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA; Departments of Biomedical Engineering, Environmental and Health Sciences, Oncology, Neurosurgery, Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Laura M Ensign
- The Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
445
|
Mortensen NP, Mercier KA, McRitchie S, Cavallo TB, Pathmasiri W, Stewart D, Sumner SJ. Microfluidics meets metabolomics to reveal the impact of Campylobacter jejuni infection on biochemical pathways. Biomed Microdevices 2016; 18:51. [PMID: 27231016 PMCID: PMC4939818 DOI: 10.1007/s10544-016-0076-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Microfluidic devices that are currently being used in pharmaceutical research also have a significant potential for utilization in investigating exposure to infectious agents. We have established a microfluidic device cultured with Caco-2 cells, and utilized metabolomics to investigate the biochemical responses to the bacterial pathogen Campylobacter jejuni. In the microfluidic devices, Caco-2 cells polarize at day 5, are uniform, have defined brush borders and tight junctions, and form a mucus layer. Metabolomics analysis of cell culture media collected from both Caco-2 cell culture systems demonstrated a more metabolic homogenous biochemical profile in the media collected from microfluidic devices, compared with media collected from transwells. GeneGo pathway mapping indicated that aminoacyl-tRNA biosynthesis was perturbed by fluid flow, suggesting that fluid dynamics and shear stress impacts the cells translational quality control. Both microfluidic device and transwell culturing systems were used to investigate the impact of Campylobacter jejuni infection on biochemical processes. Caco-2 cells cultured in either system were infected at day 5 with C. jejuni 81-176 for 48 h. Metabolomics analysis clearly differentiated C. jejuni 81-176 infected and non-infected medias collected from the microfluidic devices, and demonstrated that C. jejuni 81-176 infection in microfluidic devices impacts branched-chain amino acid metabolism, glycolysis, and gluconeogenesis. In contrast, no distinction was seen in the biochemical profiles of infected versus non-infected media collected from cells cultured in transwells. Microfluidic culturing conditions demonstrated a more metabolically homogenous cell population, and present the opportunity for studying host-pathogen interactions for extended periods of time.
Collapse
Affiliation(s)
- Ninell P Mortensen
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA.
| | - Kelly A Mercier
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Susan McRitchie
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Tammy B Cavallo
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Wimal Pathmasiri
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Delisha Stewart
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Susan J Sumner
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA.
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA.
| |
Collapse
|
446
|
Verma R, Adhikary RR, Banerjee R. Smart material platforms for miniaturized devices: implications in disease models and diagnostics. LAB ON A CHIP 2016; 16:1978-1992. [PMID: 27108534 DOI: 10.1039/c6lc00173d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Smart materials are responsive to multiple stimuli like light, temperature, pH and redox reactions with specific changes in state. Various functionalities in miniaturised devices can be achieved through the application of "smart materials" that respond to changes in their surroundings. The change in state of the materials in the presence of a stimulus may be used for on demand alteration of flow patterns in devices, acting as microvalves, as scaffolds for cellular aggregation or as modalities for signal amplification. In this review, we discuss the concepts of smart trigger responsive materials and their applications in miniaturized devices both for organ-on-a-chip disease models and for point-of-care diagnostics. The emphasis is on leveraging the smartness of these materials for example, to allow on demand sample actuation, ion dependent spheroid models for cancer or light dependent contractility of muscle films for organ-on-a-chip applications. The review throws light on the current status, scope for technological enhancements, challenges for translation and future prospects of increased incorporation of smart materials as integral parts of miniaturized devices.
Collapse
Affiliation(s)
- Ritika Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | - Rishi Rajat Adhikary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| |
Collapse
|
447
|
Marx U, Andersson TB, Bahinski A, Beilmann M, Beken S, Cassee FR, Cirit M, Daneshian M, Fitzpatrick S, Frey O, Gaertner C, Giese C, Griffith L, Hartung T, Heringa MB, Hoeng J, de Jong WH, Kojima H, Kuehnl J, Luch A, Maschmeyer I, Sakharov D, Sips AJAM, Steger-Hartmann T, Tagle DA, Tonevitsky A, Tralau T, Tsyb S, van de Stolpe A, Vandebriel R, Vulto P, Wang J, Wiest J, Rodenburg M, Roth A. Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing. ALTEX 2016; 33:272-321. [PMID: 27180100 PMCID: PMC5396467 DOI: 10.14573/altex.1603161] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023]
Abstract
The recent advent of microphysiological systems - microfluidic biomimetic devices that aspire to emulate the biology of human tissues, organs and circulation in vitro - is envisaged to enable a global paradigm shift in drug development. An extraordinary US governmental initiative and various dedicated research programs in Europe and Asia have led recently to the first cutting-edge achievements of human single-organ and multi-organ engineering based on microphysiological systems. The expectation is that test systems established on this basis would model various disease stages, and predict toxicity, immunogenicity, ADME profiles and treatment efficacy prior to clinical testing. Consequently, this technology could significantly affect the way drug substances are developed in the future. Furthermore, microphysiological system-based assays may revolutionize our current global programs of prioritization of hazard characterization for any new substances to be used, for example, in agriculture, food, ecosystems or cosmetics, thus, replacing laboratory animal models used currently. Thirty-six experts from academia, industry and regulatory bodies present here the results of an intensive workshop (held in June 2015, Berlin, Germany). They review the status quo of microphysiological systems available today against industry needs, and assess the broad variety of approaches with fit-for-purpose potential in the drug development cycle. Feasible technical solutions to reach the next levels of human biology in vitro are proposed. Furthermore, key organ-on-a-chip case studies, as well as various national and international programs are highlighted. Finally, a roadmap into the future is outlined, to allow for more predictive and regulatory-accepted substance testing on a global scale.
Collapse
|
448
|
Lu Q, Al-Sheikh O, Elisseeff JH, Grant MP. Biomaterials and Tissue Engineering Strategies for Conjunctival Reconstruction and Dry Eye Treatment. Middle East Afr J Ophthalmol 2016; 22:428-34. [PMID: 26692712 PMCID: PMC4660527 DOI: 10.4103/0974-9233.167818] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The ocular surface is a component of the anterior segment of the eye and is covered by the tear film. Together, they protect the vital external components of the eye from the environment. Injuries, surgical trauma, and autoimmune diseases can damage this system, and in severe cases, tissue engineering strategies are necessary to ensure proper wound healing and recovery. Dry eye is another major concern and a complicated disease affecting the ocular surface. More effective and innovative therapies are required for better outcomes in treating dry eye. This review focuses on the regenerative medicine of the conjunctiva, which is an essential part of the ocular surface system. Features and advances of different types of biomolecular materials, and autologous and allogeneic tissue grafts are summarized and compared. Specifically, vitrigel, a collagen membrane and novel material for use on the ocular surface, offers significant advantages over other biomaterials. This review also discusses a breakthrough microfluidic technology, “organ-on-a-chip” and its potential application in investigating new therapies for dry eye.
Collapse
Affiliation(s)
- Qiaozhi Lu
- Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA ; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Osama Al-Sheikh
- Oculoplastics and Orbit Division, King Khaled Eye Specialist Hospital, P.O. Box 7191, Riyadh 11462, Saudi Arabia
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA ; Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michael P Grant
- Oculoplastics Division, Ocular and Orbital Trauma Center, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
449
|
Xiong F, Megason SG. Abstracting the principles of development using imaging and modeling. Integr Biol (Camb) 2016; 7:633-42. [PMID: 25946995 DOI: 10.1039/c5ib00025d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Here we look at modern developmental biology with a focus on the relationship between different approaches of investigation. We argue that direct imaging is a powerful approach not only for obtaining descriptive information but also for model generation and testing that lead to mechanistic insights. Modeling, on the other hand, conceptualizes imaging data and provides guidance to perturbations. The inquiry progresses most efficiently when a trinity of approaches—quantitative imaging (measurement), modeling (theory) and perturbation (test)—are pursued in concert, but not when one approach is dominant. Using recent studies of the zebrafish system, we show how this combination has effectively advanced classic topics in developmental biology compared to a perturbation-centric approach. Finally, we show that interdisciplinary expertise and perhaps specialization are necessary for carrying out a systematic approach, and discuss the technical hurdles.
Collapse
Affiliation(s)
- Fengzhu Xiong
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
450
|
Kujala VJ, Pasqualini FS, Goss JA, Nawroth JC, Parker KK. Laminar ventricular myocardium on a microelectrode array-based chip. J Mater Chem B 2016; 4:3534-3543. [PMID: 32263387 DOI: 10.1039/c6tb00324a] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pharmaceutical screening based on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and multi electrode arrays (MEAs) have been proposed as a complementary method for electrophysiological safety and efficacy assessment in drug discovery and development. Contrary to animal models, these cells offer a human genetic background but, at present, fail to recapitulate the mechanical and structural properties of the native human myocardium. Here, we report that topographical cues on soft micromolded gelatin can coax hiPSC-CMs to form laminar cardiac tissues that resemble the native architecture of the heart. Importantly, using this method we were able to record tissue-level electrophysiological responses with a commercially available MEA setup. To validate this platform, we recorded cardiac field potentials at baseline and after pharmacological interventions with a β-adrenergic agonist (isoproterenol). Further, we tested the ability of our system to predict the response of laminar human cardiac tissues to a cardiotoxic pro-drug (terfenadine) and its non-cardiotoxic metabolite (fexofenadine). Finally, we integrated our platform with microfluidic components to build a heart-on-a-chip system that can be fluidically linked with other organs-on-chips in the future.
Collapse
Affiliation(s)
- Ville J Kujala
- Disease Biophysics Group, Harvard Stem Cell Institute, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, K.K.P. 29 Oxford Street, Pierce Hall Cambridge, MA 02130, USA.
| | | | | | | | | |
Collapse
|