1
|
Chakraborty R, Ray P, Barik S, Banik O, Mahapatra C, Banoth E, Kumar P. A Review on Microengineering of Epithelial Barriers for Biomedical and Pharmaceutical Research. ACS APPLIED BIO MATERIALS 2024; 7:8107-8125. [PMID: 39565389 DOI: 10.1021/acsabm.4c00813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Epithelial tissue forms a barrier around the human body and visceral organs, providing protection, permeation, sensation, and secretion. It is vital for our sustenance as it protects the tissue from harm and injury by restricting the entry of foreign bodies inside. Furthermore, it is a strong barrier to drugs, nutrients, and other essential deliverables. This layer also houses a large consortium of microbes, which thrive in tandem with human tissue, providing several health benefits. Moreover, the complex interplay of the microbiome with the barrier tissue is poorly understood. Therefore, replicating these barrier tissues on microdevices to generate physiological and pathophysiological models has been a huge interest for researchers over the last few decades. The artificially engineered reconstruction of these epithelial cellular barriers on microdevices could help underpin the host-microbe interaction, generating a physiological understanding of the tissue, tissue remodeling, receptor-based selective diffusion, drug testing, and others. In addition, these devices could reduce the burden of animal sacrifices for similar research and minimize the failure rate in drug discovery due to the use of primary human cells and others. This review discusses the nature of the epithelial barrier at different tissue sites, the recent developments in creating engineered barrier models, and their applications in pathophysiology, host-microbe interactions, drug discovery, and cytotoxicity. The review aims to provide know-how and knowledge behind engineered epithelial barrier tissue to bioengineers, biotechnologists, and scientists in allied fields.
Collapse
Affiliation(s)
- Ruchira Chakraborty
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Pragyan Ray
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Swagatika Barik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Oindrila Banik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Chinmaya Mahapatra
- Department of Biotechnology, National Institute of Technology, Raipur-492010 Chhattisgarh, India
| | - Earu Banoth
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Prasoon Kumar
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| |
Collapse
|
2
|
Debad S, Allen D, Bandele O, Bishop C, Blaylock M, Brown P, Bunger MK, Co JY, Crosby L, Daniel AB, Ferguson SS, Ford K, Gamboa da Costa G, Gilchrist KH, Grogg MW, Gwinn M, Hartung T, Hogan SP, Jeong YE, Kass GE, Kenyon E, Kleinstreuer NC, Kujala V, Lundquist P, Matheson J, McCullough SD, Melton-Celsa A, Musser S, Oh I, Oyetade OB, Patil SU, Petersen EJ, Sadrieh N, Sayes CM, Scruggs BS, Tan YM, Thelin B, Nelson MT, Tarazona JV, Wambaugh JF, Yang JY, Yu C, Fitzpatrick S. Trust your gut: Establishing confidence in gastrointestinal models - An overview of the state of the science and contexts of use. ALTEX 2024; 41:402-424. [PMID: 38898799 PMCID: PMC11413798 DOI: 10.14573/altex.2403261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Indexed: 06/21/2024]
Abstract
The webinar series and workshop titled “Trust Your Gut: Establishing Confidence in Gastrointestinal Models – An Overview of the State of the Science and Contexts of Use” was co-organized by NICEATM, NIEHS, FDA, EPA, CPSC, DoD, and the Johns Hopkins Center for Alternatives to Animal Testing (CAAT) and hosted at the National Institutes of Health in Bethesda, MD, USA on October 11-12, 2023. New approach methods (NAMs) for assessing issues of gastrointestinal tract (GIT)- related toxicity offer promise in addressing some of the limitations associated with animal-based assessments. GIT NAMs vary in complexity, from two-dimensional monolayer cell line-based systems to sophisticated 3-dimensional organoid systems derived from human primary cells. Despite advances in GIT NAMs, challenges remain in fully replicating the complex interactions and processes occurring within the human GIT. Presentations and discussions addressed regulatory needs, challenges, and innovations in incorporating NAMs into risk assessment frameworks; explored the state of the science in using NAMs for evaluating systemic toxicity, understanding absorption and pharmacokinetics, evaluating GIT toxicity, and assessing potential allergenicity; and discussed strengths, limitations, and data gaps of GIT NAMs as well as steps needed to establish confidence in these models for use in the regulatory setting.
Collapse
Affiliation(s)
| | | | - Omari Bandele
- Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | - Colin Bishop
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, USA
| | | | - Paul Brown
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Julia Y Co
- Complex in vitro Systems, Genentech Inc, South San Francisco, CA, USA
| | - Lynn Crosby
- Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | | | - Steve S Ferguson
- Mechanistic Toxicology Branch, Division of Translational Toxicology, NIEHS, National Institutes of Health, Bethesda, MD, USA
| | - Kevin Ford
- Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, MD, USA
| | - Gonçalo Gamboa da Costa
- FDA National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Kristin H Gilchrist
- 4D Bio³ Center for Biotechnology, Uniformed Services University, Bethesda, MD, USA
| | - Matthew W Grogg
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, USA
| | - Maureen Gwinn
- U.S. Environmental Protection Agency, Office of Research and Development, RTP, NC, USA
| | - Thomas Hartung
- Johns Hopkins University, Bloomberg School of Public Health and Whiting School of Engineering, Doerenkamp-Zbinden-Chair for Evidence-based Toxicology, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA
- University of Konstanz, CAAT-Europe, Konstanz, Germany
| | - Simon P Hogan
- Mary H. Weiser Food Allergy Center, Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ye Eun Jeong
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | - Elaina Kenyon
- U.S. Environmental Protection Agency, Office of Research and Development, RTP, NC, USA
| | | | | | | | | | | | - Angela Melton-Celsa
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Steven Musser
- Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | - Ilung Oh
- Toxicology Research Division, Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Republic of Korea
| | | | - Sarita U Patil
- Divisions of Allergy and Clinical Immunology, Departments of Medicine and Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elijah J Petersen
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Nakissa Sadrieh
- Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, Waco, TX, USA
| | | | - Yu-Mei Tan
- U.S. Environmental Protection Agency, Office of Pesticide Programs, Research Triangle Park, NC, USA
| | - Bill Thelin
- Altis Biosystems, Inc. Research Triangle Park, NC, USA
| | - M Tyler Nelson
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, USA
| | - José V Tarazona
- Spanish National Environmental Health Centre, Instituto de Salud Carlos III. Madrid, Spain
| | - John F Wambaugh
- U.S. Environmental Protection Agency, Office of Research and Development, RTP, NC, USA
| | - Jun-Young Yang
- Toxicology Research Division, Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Cheongju, Republic of Korea
| | - Changwoo Yu
- Toxicology Research Division, Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Cheongju, Republic of Korea
| | | |
Collapse
|
3
|
Baker TK, Van Vleet TR, Mahalingaiah PK, Grandhi TSP, Evers R, Ekert J, Gosset JR, Chacko SA, Kopec AK. The Current Status and Use of Microphysiological Systems by the Pharmaceutical Industry: The International Consortium for Innovation and Quality Microphysiological Systems Affiliate Survey and Commentary. Drug Metab Dispos 2024; 52:198-209. [PMID: 38123948 DOI: 10.1124/dmd.123.001510] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Microphysiological systems (MPS) are comprised of one or multiple cell types of human or animal origins that mimic the biochemical/electrical/mechanical responses and blood-tissue barrier properties of the cells observed within a complex organ. The goal of incorporating these in vitro systems is to expedite and advance the drug discovery and development paradigm with improved predictive and translational capabilities. Considering the industry need for improved efficiency and the broad challenges of model qualification and acceptance, the International Consortium for Innovation and Quality (IQ) founded an IQ MPS working group in 2014 and Affiliate in 2018. This group connects thought leaders and end users, provides a forum for crosspharma collaboration, and engages with regulators to qualify translationally relevant MPS models. To understand how pharmaceutical companies are using MPS, the IQ MPS Affiliate conducted two surveys in 2019, survey 1, and 2021, survey 2, which differed slightly in the scope of definition of the complex in vitro models under question. The surveys captured demographics, resourcing, rank order for organs of interest, compound modalities tested, and MPS organ-specific questions, including nonclinical species needs and cell types. The major focus of this manuscript is on results from survey 2, where we specifically highlight the context of use for MPS within safety, pharmacology, or absorption, disposition, metabolism, and excretion and discuss considerations for including MPS data in regulatory submissions. In summary, these data provide valuable insights for developers, regulators, and pharma, offering a view into current industry practices and future considerations while highlighting key challenges impacting MPS adoption. SIGNIFICANCE STATEMENT: The application of microphysiological systems (MPS) represents a growing area of interest in the drug discovery and development framework. This study surveyed 20+ pharma companies to understand resourcing, current areas of application, and the key challenges and barriers to internal MPS adoption. These results will provide regulators, tech providers, and pharma industry leaders a starting point to assess the current state of MPS applications along with key learnings to effectively realize the potential of MPS as an emerging technology.
Collapse
Affiliation(s)
- Thomas K Baker
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.) baker_thomas_k@lilly
| | - Terry R Van Vleet
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Prathap Kumar Mahalingaiah
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Taraka Sai Pavan Grandhi
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Raymond Evers
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Jason Ekert
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - James R Gosset
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Silvi A Chacko
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| | - Anna K Kopec
- Investigative Toxicology, Eli Lilly, Indianapolis, Indiana (T.K.B.); Investigative Toxicology and Pathology, AbbVie, Inc., Chicago, Illinois (T.R.V.F., P.K.M.); Complex In Vitro Models Group, GSK, Collegeville, Pennsylvania (T.S.P.G.); Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania (R.E.); UCB Pharma, Cambridge, Massachusetts (J.E.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc., Cambridge, Massachusetts (J.R.G.); Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey (S.A.C.); and Drug Safety Research & Development, Pfizer, Inc., Groton, Connecticut (A.K.K.)
| |
Collapse
|
4
|
Xian C, Zhang J, Zhao S, Li XG. Gut-on-a-chip for disease models. J Tissue Eng 2023; 14:20417314221149882. [PMID: 36699635 PMCID: PMC9869227 DOI: 10.1177/20417314221149882] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
The intestinal tract is a vital organ responsible for digestion and absorption in the human body and plays an essential role in pathogen invasion. Compared with other traditional models, gut-on-a-chip has many unique advantages, and thereby, it can be considered as a novel model for studying intestinal functions and diseases. Based on the chip design, we can replicate the in vivo microenvironment of the intestine and study the effects of individual variables on the experiment. In recent years, it has been used to study several diseases. To better mimic the intestinal microenvironment, the structure and function of gut-on-a-chip are constantly optimised and improved. Owing to the complexity of the disease mechanism, gut-on-a-chip can be used in conjunction with other organ chips. In this review, we summarise the human intestinal structure and function as well as the development and improvement of gut-on-a-chip. Finally, we present and discuss gut-on-a-chip applications in inflammatory bowel disease (IBD), viral infections and phenylketonuria. Further improvement of the simulation and high throughput of gut-on-a-chip and realisation of personalised treatments are the problems that should be solved for gut-on-a-chip as a disease model.
Collapse
Affiliation(s)
| | | | | | - Xiang-Guang Li
- Xiang-Guang Li, Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, No. 100 Waihuan Xi Road (GDUT), Panyu District, Guangzhou 510006, China.
| |
Collapse
|
5
|
Jiang L, Li Q, Liang W, Du X, Yang Y, Zhang Z, Xu L, Zhang J, Li J, Chen Z, Gu Z. Organ-On-A-Chip Database Revealed-Achieving the Human Avatar in Silicon. Bioengineering (Basel) 2022; 9:685. [PMID: 36421086 PMCID: PMC9687773 DOI: 10.3390/bioengineering9110685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Organ-on-a-chip (OOC) provides microphysiological conditions on a microfluidic chip, which makes up for the shortcomings of traditional in vitro cellular culture models and animal models. It has broad application prospects in drug development and screening, toxicological mechanism research, and precision medicine. A large amount of data could be generated through its applications, including image data, measurement data from sensors, ~omics data, etc. A database with proper architecture is required to help scholars in this field design experiments, organize inputted data, perform analysis, and promote the future development of novel OOC systems. In this review, we overview existing OOC databases that have been developed, including the BioSystics Analytics Platform (BAP) developed by the University of Pittsburgh, which supports study design as well as data uploading, storage, visualization, analysis, etc., and the organ-on-a-chip database (Ocdb) developed by Southeast University, which has collected a large amount of literature and patents as well as relevant toxicological and pharmaceutical data and provides other major functions. We used examples to overview how the BAP database has contributed to the development and applications of OOC technology in the United States for the MPS consortium and how the Ocdb has supported researchers in the Chinese Organoid and Organs-On-A-Chip society. Lastly, the characteristics, advantages, and limitations of these two databases were discussed.
Collapse
Affiliation(s)
- Lincao Jiang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Weicheng Liang
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Xuan Du
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Yi Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Zilin Zhang
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Lili Xu
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Jing Zhang
- Jiangsu Avartarget Biotechnology Corp., Suzhou 215163, China
| | - Jian Li
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, SiPaiLou # 2, Nanjing 210096, China
| |
Collapse
|
6
|
Smith EM, Grassel CL, Papadimas A, Foulke-Abel J, Barry EM. The role of CFA/I in adherence and toxin delivery by ETEC expressing multiple colonization factors in the human enteroid model. PLoS Negl Trop Dis 2022; 16:e0010638. [PMID: 35881640 PMCID: PMC9355178 DOI: 10.1371/journal.pntd.0010638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/05/2022] [Accepted: 07/07/2022] [Indexed: 01/10/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a primary causative agent of diarrhea in travelers and young children in low-to-middle-income countries (LMICs). ETEC adhere to intestinal epithelia via colonization factors (CFs) and secrete heat-stable toxin (ST) and/or heat-labile toxin (LT), causing dysregulated cellular ion transport and water secretion. ETEC isolates often harbor genes encoding more than one CF that are targets as vaccine antigens. CFA/I is a major CF that is associated with ETEC that causes moderate-to-severe diarrhea and plays an important role in pathogenesis. The Global Enteric Multicenter Study finding that 78% of CFA/I-expressing ETEC also encode the minor CF CS21 prompted investigation of the combined role of these two CFs. Western blots and electron microscopy demonstrated growth media-dependent and strain-dependent differences in CFA/I and CS21 expression. The critical role of CFA/I in adherence by ETEC strains expressing CFA/I and CS21 was demonstrated using the human enteroid model and a series of CFA/I- and CS21-specific mutants. Furthermore, only anti-CFA/I antibodies inhibited adherence by global ETEC isolates expressing CFA/I and CS21. Delivery of ST and resulting cGMP secretion was measured in supernatants from infected enteroid monolayers, and strain-specific ST delivery and time-dependent cGMP production was observed. Interestingly, cGMP levels were similar across wildtype and CF-deficient strains, reflecting a limitation of this static aerobic infection model. Despite adherence by ETEC and delivery of ST, the enteroid monolayer integrity was not disrupted, as shown by the lack of decrease in transepithelial electrical resistance and the lack of IL-8 cytokines produced during infection. Taken together, these data demonstrate that targeting CFA/I in global clinical CFA/I-CS21 strains is sufficient for adherence inhibition, supporting a vaccine strategy that focuses on blocking major CFs. In addition, the human enteroid model has significant utility for the study of ETEC pathogenesis and evaluation of vaccine-induced functional antibody responses.
Collapse
Affiliation(s)
- Emily M. Smith
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Christen L. Grassel
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Antonia Papadimas
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer Foulke-Abel
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Eileen M. Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
7
|
Epithelial and Neutrophil Interactions and Coordinated Response to Shigella in a Human Intestinal Enteroid-Neutrophil Coculture Model. mBio 2022; 13:e0094422. [PMID: 35652591 PMCID: PMC9239269 DOI: 10.1128/mbio.00944-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Polymorphonuclear neutrophils (PMN) are recruited to the gastrointestinal mucosa in response to inflammation, injury, and infection. Here, we report the development and the characterization of an ex vivo tissue coculture model consisting of human primary intestinal enteroid monolayers and PMN, and a mechanistic interrogation of PMN-epithelial cell interaction and response to Shigella, a primary cause of childhood dysentery. Cellular adaptation and tissue integration, barrier function, PMN phenotypic and functional attributes, and innate immune responses were examined. PMN within the enteroid monolayers acquired a distinct activated/migratory phenotype that was influenced by direct epithelial cell contact as well as by molecular signals. Seeded on the basal side of the intestinal monolayer, PMN were intercalated within the epithelial cells and moved paracellularly toward the apical side. Cocultured PMN also increased basal secretion of interleukin 8 (IL-8). Shigella added to the apical surface of the monolayers evoked additional PMN phenotypic adaptations, including increased expression of cell surface markers associated with chemotaxis and cell degranulation (CD47, CD66b, and CD88). Apical Shigella infection triggered rapid transmigration of PMN to the luminal side, neutrophil extracellular trap (NET) formation, and bacterial phagocytosis and killing. Shigella infection modulated cytokine production in the coculture; apical monocyte chemoattractant protein (MCP-1), tumor necrosis factor alpha (TNF-α), and basolateral IL-8 production were downregulated, while basolateral IL-6 secretion was increased. We demonstrated, for the first time, PMN phenotypic adaptation and mobilization and coordinated epithelial cell-PMN innate response upon Shigella infection in the human intestinal environment. The enteroid monolayer-PMN coculture represents a technical innovation for mechanistic interrogation of gastrointestinal physiology, host-microbe interaction, innate immunity, and evaluation of preventive/therapeutic tools.
Collapse
|
8
|
Fernandes J, Karra N, Bowring J, Reale R, James J, Blume C, Pell TJ, Rowan WC, Davies DE, Swindle EJ, Morgan H. Real-time monitoring of epithelial barrier function by impedance spectroscopy in a microfluidic platform. LAB ON A CHIP 2022; 22:2041-2054. [PMID: 35485428 DOI: 10.1039/d1lc01046h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
A multichannel microfluidic platform for real-time monitoring of epithelial barrier integrity by electrical impedance has been developed. Growth and polarization of human epithelial cells from the airway or gastrointestinal tract was continuously monitored over 5 days in 8 parallel, individually perfused microfluidic chips. Electrical impedance data were continuously recorded to monitor cell barrier formation using a low-cost bespoke impedance analyser. Data was analysed using an electric circuit model to extract the equivalent transepithelial electrical resistance and epithelial cell layer capacitance. The cell barrier integrity steadily increased overtime, achieving an average resistance of 418 ± 121 Ω cm2 (airway cells) or 207 ± 59 Ω cm2 (gastrointestinal cells) by day 5. The utility of the polarized airway epithelial barrier was demonstrated using a 24 hour challenge with double stranded RNA to mimic viral infection. This caused a rapid decrease in barrier integrity in association with disruption of tight junctions, whereas simultaneous treatment with a corticosteroid reduced this effect. The platform is able to measure barrier integrity in real-time and is scalable, thus has the potential to be used for drug development and testing.
Collapse
Affiliation(s)
- João Fernandes
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, UK.
| | - Nikita Karra
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, UK.
| | - Joel Bowring
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, UK.
| | - Riccardo Reale
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, UK.
| | - Jonathan James
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Cornelia Blume
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
- Institute for Life Sciences, University of Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK
| | - Theresa J Pell
- Novel Human Genetics Research Unit, GlaxoSmithKline R&D, Stevenage, Hertfordshire, UK
| | - Wendy C Rowan
- Novel Human Genetics Research Unit, GlaxoSmithKline R&D, Stevenage, Hertfordshire, UK
| | - Donna E Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
- Institute for Life Sciences, University of Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK
| | - Emily J Swindle
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
- Institute for Life Sciences, University of Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK
| | - Hywel Morgan
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, UK.
- Institute for Life Sciences, University of Southampton, UK
| |
Collapse
|
9
|
Arian CM, Imaoka T, Yang J, Kelly EJ, Thummel KE. Gutsy science: In vitro systems of the human intestine to model oral drug disposition. Pharmacol Ther 2022; 230:107962. [PMID: 34478775 PMCID: PMC8821120 DOI: 10.1016/j.pharmthera.2021.107962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 02/03/2023]
Abstract
The intestine has important gate-keeping functions that can profoundly affect the systemic blood exposure of orally administered drugs. Thus, characterizing a new molecular entity's (NME) disposition within the intestine is of utmost importance in drug development. While currently used in vitro systems, such as Ussing chamber, precision-cut intestinal slices, immortalized cell lines, and primary enterocytes provide substantial knowledge about drug absorption and the intestinal first-pass effect, they remain sub-optimal for quantitatively predicting this process and the oral bioavailability of many drugs. Use of novel in vitro systems such as intestinal organoids and intestinal microphysiological systems have provided substantial advances over the past decade, expanding our understanding of intestinal physiology, pathology, and development. However, application of these emerging in vitro systems in the pharmaceutical science is in its infancy. Preliminary work has demonstrated that these systems more accurately recapitulate the physiology and biochemistry of the intact intestine, as it relates to oral drug disposition, and thus they hold considerable promise as preclinical testing platforms of the future. Here we review currently used and emerging in vitro models of the human intestine employed in pharmaceutical science research. We also highlight aspects of these emerging tools that require further study.
Collapse
Affiliation(s)
- Christopher M Arian
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Tomoki Imaoka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Jade Yang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
10
|
Nguyen OTP, Misun PM, Lohasz C, Lee J, Wang W, Schroeder T, Hierlemann A. An Immunocompetent Microphysiological System to Simultaneously Investigate Effects of Anti-Tumor Natural Killer Cells on Tumor and Cardiac Microtissues. Front Immunol 2021; 12:781337. [PMID: 34925361 PMCID: PMC8675866 DOI: 10.3389/fimmu.2021.781337] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/16/2021] [Indexed: 12/26/2022] Open
Abstract
Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Patrick M. Misun
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Christian Lohasz
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jihyun Lee
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Weijia Wang
- Cell Systems Dynamics Group, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Timm Schroeder
- Cell Systems Dynamics Group, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
11
|
Franco YL, Da Silva L, Cristofoletti R. Navigating Through Cell-Based In vitro Models Available for Prediction of Intestinal Permeability and Metabolism: Are We Ready for 3D? AAPS J 2021; 24:2. [PMID: 34811603 PMCID: PMC8925318 DOI: 10.1208/s12248-021-00665-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/23/2021] [Indexed: 01/07/2023] Open
Abstract
Traditionally, in vitro studies to quantify the
intestinal permeability of drugs have relied on two-dimensional cell culture models using human colorectal carcinoma cell lines, namely Caco-2, HT 29 and T84 cells. Although these models have been commonly used for high-throughput screening of xenobiotics in preclinical studies, they do not fully recapitulate the morphology and functionality of enterocytes found in the human intestine in vivo. Efforts to improve the physiological and functional relevance of in vitro intestinal models have led to the development of enteroids/intestinal organoids and microphysiological systems. These models leverage advances in three-dimensional cell culture techniques and stem cell technology (in addition to microfluidics for microphysiological systems), to mimic the architecture and microenvironment of the in vivo intestine more accurately. In this commentary, we will discuss the advantages and limitations of these established and emerging intestinal models, as well as their current and potential future applications for the pre-clinical assessment of oral therapies.
Collapse
Affiliation(s)
- Yesenia L Franco
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Lais Da Silva
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA.
| |
Collapse
|
12
|
Rodrigues DB, Failla ML. Intestinal cell models for investigating the uptake, metabolism and absorption of dietary nutrients and bioactive compounds. Curr Opin Food Sci 2021. [DOI: 10.1016/j.cofs.2021.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
13
|
Swaminathan G, Kamyabi N, Carter HE, Rajan A, Karandikar U, Criss ZK, Shroyer NF, Robertson MJ, Coarfa C, Huang C, Shannon TE, Tadros M, Estes MK, Maresso AW, Grande-Allen KJ. Effect of substrate stiffness on human intestinal enteroids' infectivity by enteroaggregative Escherichia coli. Acta Biomater 2021; 132:245-259. [PMID: 34280559 PMCID: PMC8434991 DOI: 10.1016/j.actbio.2021.07.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/24/2021] [Accepted: 07/12/2021] [Indexed: 01/08/2023]
Abstract
Human intestinal enteroids (HIE) models have contributed significantly to our understanding of diarrheal diseases and other intestinal infections, but their routine culture conditions fail to mimic the mechanical environment of the native intestinal wall. Because the mechanical characteristics of the intestine significantly alter how pathogens interact with the intestinal epithelium, we used different concentrations of polyethylene glycol (PEG) to generate soft (~2 kPa), medium (~10 kPa), and stiff (~100 kPa) hydrogel biomaterial scaffolds. The height of HIEs cultured in monolayers atop these hydrogels was 18 µm whereas HIEs grown on rigid tissue culture surfaces (with stiffness in the GPa range) were 10 µm. Substrate stiffness also influenced the amount of enteroaggregative E. coli (EAEC strain 042) adhered to the HIEs. We quantified a striking difference in adherence pattern; on the medium and soft gels, the bacteria formed clusters of > 100 and even > 1000 on both duodenal and jejunal HIEs (such as would be found in biofilms), but did not on glass slides and stiff hydrogels. All hydrogel cultured HIEs showed significant enrichment for gene and signaling pathways related to epithelial differentiation, cell junctions and adhesions, extracellular matrix, mucins, and cell signaling compared to the HIEs cultured on rigid tissue culture surfaces. Collectively, these results indicate that the HIE monolayers cultured on the hydrogels are primed for a robust engagement with their mechanical environment, and that the soft hydrogels promote the formation of larger EAEC aggregates, likely through an indirect differential effect on mucus. STATEMENT OF SIGNIFICANCE: Enteroids are a form of in vitro experimental mini-guts created from intestinal stem cells. Enteroids are usually cultured in 3D within Matrigel atop rigid glass or plastic substrates, which fail to mimic the native intestinal mechanical environment. Because intestinal mechanics significantly alter how pathogens interact with the intestinal epithelium, we grew human intestinal enteroids in 2D atop polyethylene glycol (PEG) hydrogel scaffolds that were soft, medium, or stiff. Compared with enteroids grown in 2D atop glass or plastic, the enteroids grown on hydrogels were taller and more enriched in mechanobiology-related gene signaling pathways. Additionally, enteroids on the softest hydrogels supported adhesion of large aggregates of enteroaggregative E. coli. Thus, this platform offers a more biomimetic model for studying enteric diseases.
Collapse
Affiliation(s)
- Ganesh Swaminathan
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States
| | - Nabiollah Kamyabi
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States
| | - Hannah E Carter
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Umesh Karandikar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Zachary K Criss
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States
| | - Noah F Shroyer
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States
| | - Matthew J Robertson
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Cristian Coarfa
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States; Department of Molecular and Cellular Biology - Molecular Regulation, Baylor College of Medicine, Houston, TX, United States
| | - Chenlin Huang
- Department of Biosciences, Rice University, Houston, TX, United States
| | - Tate E Shannon
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States
| | - Madeleine Tadros
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, United States
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Anthony W Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - K Jane Grande-Allen
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States.
| |
Collapse
|
14
|
Ramadan Q, Fardous RS, Hazaymeh R, Alshmmari S, Zourob M. Pharmacokinetics-On-a-Chip: In Vitro Microphysiological Models for Emulating of Drugs ADME. Adv Biol (Weinh) 2021; 5:e2100775. [PMID: 34323392 DOI: 10.1002/adbi.202100775] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Despite many ongoing efforts across the full spectrum of pharmaceutical and biotech industries, drug development is still a costly undertaking that involves a high risk of failure during clinical trials. Animal models played vital roles in understanding the mechanism of human diseases. However, the use of these models has been a subject of heated debate, particularly due to ethical matters and the inevitable pathophysiological differences between animals and humans. Current in vitro models lack the sufficient functionality and predictivity of human pharmacokinetics and toxicity, therefore, are not capable to fully replace animal models. The recent development of micro-physiological systems has shown great potential as indispensable tools for recapitulating key physiological parameters of humans and providing in vitro methods for predicting the pharmacokinetics and pharmacodynamics in humans. Integration of Absorption, Distribution, Metabolism, and Excretion (ADME) processes within one close in vitro system is a paramount development that would meet important unmet pharmaceutical industry needs. In this review paper, synthesis of the ADME-centered organ-on-a-chip technology is systemically presented from what is achieved to what needs to be done, emphasizing the requirements of in vitro models that meet industrial needs in terms of the structure and functions.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Roa Saleem Fardous
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia.,Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, G4 0RE, United Kingdom
| | - Rana Hazaymeh
- Almaarefa University, Riyadh, 13713, Kingdom of Saudi Arabia
| | - Sultan Alshmmari
- Saudi Food and Drug Authority, Riyadh, 13513-7148, Kingdom of Saudi Arabia
| | | |
Collapse
|
15
|
Wilson RL, Hewes SA, Rajan A, Lin SC, Bomidi C, Iida T, Estes MK, Maresso AW, Grande-Allen KJ. A Millifluidic Perfusion Cassette for Studying the Pathogenesis of Enteric Infections Using Ex-Vivo Organoids. Ann Biomed Eng 2021; 49:1233-1244. [PMID: 33409849 PMCID: PMC8012234 DOI: 10.1007/s10439-020-02705-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
To generate physiologically-relevant experimental models, the study of enteric diarrheal diseases is turning increasingly to advanced in vitro models that combine ex vivo, stem cell-derived "organoid" cell lines with bioengineered culture environments that expose them to mechanical stimuli, such as fluid flow. However, such approaches require considerable technical expertise with both microfabrication and organoid culture, and are, therefore, inaccessible to many researchers. For this reason, we have developed a perfusion system that is simple to fabricate, operate, and maintain. Its dimensions approximate the volume and cell culture area of traditional 96-well plates and allow the incorporation of fastidious primary, stem cell-derived cell lines with only minimal adaptation of their established culture techniques. We show that infections with enteroaggregative E. coli and norovirus, common causes of infectious diarrhea, in the system display important differences from static models, and in some ways better recreate the pathophysiology of in vivo infections. Furthermore, commensal strains of bacteria can be added alongside the pathogens to simulate the effects of a host microbiome on the infectious process. For these reasons, we believe that this perfusion system is a powerful, yet easily accessible tool for studying host-pathogen interactions in the human intestine.
Collapse
Affiliation(s)
- Reid L Wilson
- Department of Bioengineering, Rice University, 6100 Main St, MS-142, Houston, TX, 77005, USA
- Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sarah A Hewes
- Department of Bioengineering, Rice University, 6100 Main St, MS-142, Houston, TX, 77005, USA
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Shih-Ching Lin
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Takanori Iida
- Department of Bioengineering, Rice University, 6100 Main St, MS-142, Houston, TX, 77005, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Anthony W Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - K Jane Grande-Allen
- Department of Bioengineering, Rice University, 6100 Main St, MS-142, Houston, TX, 77005, USA.
| |
Collapse
|
16
|
Alexander KL, Serrano CA, Chakraborty A, Nearing M, Council LN, Riquelme A, Garrido M, Bellis SL, Smythies LE, Smith PD. Modulation of glycosyltransferase ST6Gal-I in gastric cancer-derived organoids disrupts homeostatic epithelial cell turnover. J Biol Chem 2020; 295:14153-14163. [PMID: 32763973 DOI: 10.1074/jbc.ra120.014887] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/05/2020] [Indexed: 12/24/2022] Open
Abstract
Programmed cell death promotes homeostatic cell turnover in the epithelium but is dysregulated in cancer. The glycosyltransferase ST6Gal-I is known to block homeostatic apoptosis through α2,6-linked sialylation of the death receptor TNFR1 in many cell types. However, its role has not been investigated in gastric epithelial cells or gastric tumorigenesis. We determined that human gastric antral epithelium rarely expressed ST6Gal-I, but the number of ST6Gal-I-expressing epithelial cells increased significantly with advancing premalignancy leading to cancer. The mRNA expression levels of ST6GAL-I and SOX9 in human gastric epithelial cells correlated positively with one another through the premalignancy cascade, indicating that increased epithelial cell expression of ST6Gal-I is associated with premalignant progression. To determine the functional impact of increased ST6Gal-I, we generated human gastric antral organoids from epithelial stem cells and differentiated epithelial monolayers from gastric organoids. Gastric epithelial stem cells strongly expressed ST6Gal-I, suggesting a novel biomarker of stemness. In contrast, organoid-derived epithelial monolayers expressed markedly reduced ST6Gal-I and underwent TNF-induced, caspase-mediated apoptosis, consistent with homeostasis. Conversely, epithelial monolayers generated from gastric cancer stem cells retained high levels of ST6Gal-I and resisted TNF-induced apoptosis, supporting prolonged survival. Protection from TNF-induced apoptosis depended on ST6Gal-I overexpression, because forced ST6Gal-I overexpression in normal gastric stem cell-differentiated monolayers inhibited TNF-induced apoptosis, and cleavage of α2,6-linked sialic acids from gastric cancer organoid-derived monolayers restored susceptibility to TNF-induced apoptosis. These findings implicate up-regulated ST6Gal-I expression in blocking homeostatic epithelial cell apoptosis in gastric cancer pathogenesis, suggesting a mechanism for prolonged epithelioid tumor cell survival.
Collapse
Affiliation(s)
- Katie L Alexander
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Carolina A Serrano
- Departments of Pediatric Gastroenterology and Nutrition, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Asmi Chakraborty
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marie Nearing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Leona N Council
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Arnoldo Riquelme
- Department of Gastroenterologys, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Department Health Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Marcelo Garrido
- Department of Oncology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Susan L Bellis
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lesley E Smythies
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Phillip D Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
17
|
Ayechu-Muruzabal V, Overbeek SA, Kostadinova AI, Stahl B, Garssen J, van’t Land B, Willemsen LE. Exposure of Intestinal Epithelial Cells to 2'-Fucosyllactose and CpG Enhances Galectin Release and Instructs Dendritic Cells to Drive Th1 and Regulatory-Type Immune Development. Biomolecules 2020; 10:E784. [PMID: 32438601 PMCID: PMC7278199 DOI: 10.3390/biom10050784] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 05/15/2020] [Indexed: 01/01/2023] Open
Abstract
Intestinal epithelial cells (IEC) release immunomodulatory galectins upon exposure to CpG DNA (mimicking bacterial triggers) and short-chain galacto- and long-chain fructo-oligosaccharides (GF). This study aims to investigate the immunomodulatory properties of 2'-fucosyllactose (2'-FL), a non-digestible oligosaccharide (NDO) abundantly present in human milk, using a co-culture model developed to study the crosstalk between IEC and innate and adaptive immune cells. IECs, co-cultured with αCD3/CD28-activated peripheral blood mononuclear cells (PBMC), were apically exposed to NDOs and CpG, washed and co-cultured with immature monocyte-derived dendritic cells (moDC). Subsequently, moDC were co-cultured with naïve CD4+ T-cells. In the presence of CpG, both 2'-FL or GF-exposed IEC enhanced Th1-type IFNγ and regulatory IL-10 secretion of PBMCs, compared to CpG alone, while Th2-type IL-13 was reduced. Both NDOs increased IEC-derived galectin-3, -4, -9 and TGF-β1 of CpG-exposed IEC. Only galectin-9 correlated with all modified immune parameters and TGF-β1 secretion. MoDCs exposed to 2'-FL and CpG-conditioned IEC instructed IFNγ and IL-10 secretion by CD4+ T-cells, suggesting the development of a regulatory Th1 response. These results reveal that 2'-FL and GF could contribute to the mucosal immune development by supporting the effect of microbial CpG DNA associated with the modulation of epithelial galectin and TGF-β1 secretion.
Collapse
Affiliation(s)
- Veronica Ayechu-Muruzabal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (V.A.-M.); (J.G.)
| | - Saskia A. Overbeek
- Global Centre of Excellence in Immunology, Danone Nutricia Research B.V., 3584 CT Utrecht, The Netherlands; (S.A.O.); (A.I.K.); (B.v.L.)
| | - Atanaska I. Kostadinova
- Global Centre of Excellence in Immunology, Danone Nutricia Research B.V., 3584 CT Utrecht, The Netherlands; (S.A.O.); (A.I.K.); (B.v.L.)
| | - Bernd Stahl
- Global Centre of Excellence in Human Milk Research & Analytical Sciences, Danone Nutricia Research B.V., 3584 CT Utrecht, The Netherlands;
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (V.A.-M.); (J.G.)
- Global Centre of Excellence in Immunology, Danone Nutricia Research B.V., 3584 CT Utrecht, The Netherlands; (S.A.O.); (A.I.K.); (B.v.L.)
| | - Belinda van’t Land
- Global Centre of Excellence in Immunology, Danone Nutricia Research B.V., 3584 CT Utrecht, The Netherlands; (S.A.O.); (A.I.K.); (B.v.L.)
- Center for Translational Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Linette E.M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (V.A.-M.); (J.G.)
| |
Collapse
|
18
|
Peters MF, Choy AL, Pin C, Leishman DJ, Moisan A, Ewart L, Guzzie-Peck PJ, Sura R, Keller DA, Scott CW, Kolaja KL. Developing in vitro assays to transform gastrointestinal safety assessment: potential for microphysiological systems. LAB ON A CHIP 2020; 20:1177-1190. [PMID: 32129356 DOI: 10.1039/c9lc01107b] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Drug-induced gastrointestinal toxicities (DI-GITs) are among the most common adverse events in clinical trials. High prevalence of DI-GIT has persisted among new drugs due in part to the lack of robust experimental tools to allow early detection or to guide optimization of safer molecules. Developing in vitro assays for the leading GI toxicities (nausea, vomiting, diarrhoea, constipation, and abdominal pain) will likely involve recapitulating complex physiological properties that require contributions from diverse cell/tissue types including epithelial, immune, microbiome, nerve, and muscle. While this stipulation may be beyond traditional 2D monocultures of intestinal cell lines, emerging 3D GI microtissues capture interactions between diverse cell and tissue types. These interactions give rise to microphysiologies fundamental to gut biology. For GI microtissues, organoid technology was the breakthrough that introduced intestinal stem cells with the capability of differentiating into each of the epithelial cell types and that self-organize into a multi-cellular tissue proxy with villus- and crypt-like domains. Recently, GI microtissues generated using miniaturized devices with microfluidic flow and cyclic peristaltic strain were shown to induce Caco2 cells to spontaneously differentiate into each of the principle intestinal epithelial cell types. Second generation models comprised of epithelial organoids or microtissues co-cultured with non-epithelial cell types can successfully reproduce cross-'tissue' functional interactions broadening the potential of these models to accurately study drug-induced toxicities. A new paradigm in which in vitro assays become an early part of GI safety assessment could be realized if microphysiological systems (MPS) are developed in alignment with drug-discovery needs. Herein, approaches for assessing GI toxicity of pharmaceuticals are reviewed and gaps are compared with capabilities of emerging GI microtissues (e.g., organoids, organ-on-a-chip, transwell systems) in order to provide perspective on the assay features needed for MPS models to be adopted for DI-GIT assessment.
Collapse
Affiliation(s)
- Matthew F Peters
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Peters MF, Landry T, Pin C, Maratea K, Dick C, Wagoner MP, Choy AL, Barthlow H, Snow D, Stevens Z, Armento A, Scott CW, Ayehunie S. Human 3D Gastrointestinal Microtissue Barrier Function As a Predictor of Drug-Induced Diarrhea. Toxicol Sci 2020; 168:3-17. [PMID: 30364994 PMCID: PMC6390652 DOI: 10.1093/toxsci/kfy268] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Drug-induced gastrointestinal toxicities (GITs) rank among the most common clinical side effects. Preclinical efforts to reduce incidence are limited by inadequate predictivity of in vitro assays. Recent breakthroughs in in vitro culture methods support intestinal stem cell maintenance and continual differentiation into the epithelial cell types resident in the intestine. These diverse cells self-assemble into microtissues with in vivo-like architecture. Here, we evaluate human GI microtissues grown in transwell plates that allow apical and/or basolateral drug treatment and 96-well throughput. Evaluation of assay utility focused on predictivity for diarrhea because this adverse effect correlates with intestinal barrier dysfunction which can be measured in GI microtissues using transepithelial electrical resistance (TEER). A validation set of widely prescribed drugs was assembled and tested for effects on TEER. When the resulting TEER inhibition potencies were adjusted for clinical exposure, a threshold was identified that distinguished drugs that induced clinical diarrhea from those that lack this liability. Microtissue TEER assay predictivity was further challenged with a smaller set of drugs whose clinical development was limited by diarrhea that was unexpected based on 1-month animal studies. Microtissue TEER accurately predicted diarrhea for each of these drugs. The label-free nature of TEER enabled repeated quantitation with sufficient precision to develop a mathematical model describing the temporal dynamics of barrier damage and recovery. This human 3D GI microtissue is the first in vitro assay with validated predictivity for diarrhea-inducing drugs. It should provide a platform for lead optimization and offers potential for dose schedule exploration.
Collapse
Affiliation(s)
- Matthew F Peters
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Tim Landry
- MatTek Corporation, Ashland, Massachusetts 01721
| | - Carmen Pin
- Mechanistic Safety and ADME Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Kim Maratea
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Cortni Dick
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Matthew P Wagoner
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Allison L Choy
- Science and Enabling Units IT, AstraZeneca, Waltham, MA 02451
| | - Herb Barthlow
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Deb Snow
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | | | - Alex Armento
- MatTek Corporation, Ashland, Massachusetts 01721
| | - Clay W Scott
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | | |
Collapse
|
20
|
Fowler S, Chen WLK, Duignan DB, Gupta A, Hariparsad N, Kenny JR, Lai WG, Liras J, Phillips JA, Gan J. Microphysiological systems for ADME-related applications: current status and recommendations for system development and characterization. LAB ON A CHIP 2020; 20:446-467. [PMID: 31932816 DOI: 10.1039/c9lc00857h] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharma Research and Early Development, F.Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | | | - David B Duignan
- Department of Drug Metabolism, Pharmacokinetics & Bioanalysis, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, USA
| | - Anshul Gupta
- Amgen Research, 360 Binney St, Cambridge, MA 02141, USA
| | - Niresh Hariparsad
- Department of Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, USA
| | - Jane R Kenny
- DMPK, Genentech, 1 DNA Way, South San Francisco 94080, USA
| | | | - Jennifer Liras
- Medicine Design, Pfizer Inc, 1 Portland Ave, Cambridge, MA 02139, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb R&D, PO Box 4000, Princeton, NJ 08543-4000, USA.
| |
Collapse
|
21
|
Isoherranen N, Madabushi R, Huang S. Emerging Role of Organ-on-a-Chip Technologies in Quantitative Clinical Pharmacology Evaluation. Clin Transl Sci 2019; 12:113-121. [PMID: 30740886 PMCID: PMC6440571 DOI: 10.1111/cts.12627] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/26/2019] [Indexed: 12/28/2022] Open
Abstract
The recently enacted Prescription Drug User Fee Act (PDUFA) VI includes in its performance goals "enhancing regulatory science and expediting drug development." The key elements in "enhancing regulatory decision tools to support drug development and review" include "advancing model-informed drug development (MIDD)." This paper describes (i) the US Food and Drug Administration (FDA) Office of Clinical Pharmacology's continuing efforts in developing quantitative clinical pharmacology models (disease, drug, and clinical trial models) to advance MIDD, (ii) how emerging novel tools, such as organ-on-a-chip technologies or microphysiological systems, can provide new insights into physiology and disease mechanisms, biomarker identification and evaluation, and elucidation of mechanisms of adverse drug reactions, and (iii) how the single organ or linked organ microphysiological systems can provide critical system parameters for improved physiologically-based pharmacokinetic and pharmacodynamic evaluations. Continuous public-private partnerships are critical to advance this field and in the application of these new technologies in drug development and regulatory review.
Collapse
Affiliation(s)
- Nina Isoherranen
- Office of Clinical Pharmacology (OCP)Office of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
- Department of PharmaceuticsSchool of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Rajanikanth Madabushi
- Office of Clinical Pharmacology (OCP)Office of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Shiew‐Mei Huang
- Office of Clinical Pharmacology (OCP)Office of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug Administration (FDA)Silver SpringMarylandUSA
| |
Collapse
|
22
|
Taylor DL, Gough A, Schurdak ME, Vernetti L, Chennubhotla CS, Lefever D, Pei F, Faeder JR, Lezon TR, Stern AM, Bahar I. Harnessing Human Microphysiology Systems as Key Experimental Models for Quantitative Systems Pharmacology. Handb Exp Pharmacol 2019; 260:327-367. [PMID: 31201557 PMCID: PMC6911651 DOI: 10.1007/164_2019_239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Two technologies that have emerged in the last decade offer a new paradigm for modern pharmacology, as well as drug discovery and development. Quantitative systems pharmacology (QSP) is a complementary approach to traditional, target-centric pharmacology and drug discovery and is based on an iterative application of computational and systems biology methods with multiscale experimental methods, both of which include models of ADME-Tox and disease. QSP has emerged as a new approach due to the low efficiency of success in developing therapeutics based on the existing target-centric paradigm. Likewise, human microphysiology systems (MPS) are experimental models complementary to existing animal models and are based on the use of human primary cells, adult stem cells, and/or induced pluripotent stem cells (iPSCs) to mimic human tissues and organ functions/structures involved in disease and ADME-Tox. Human MPS experimental models have been developed to address the relatively low concordance of human disease and ADME-Tox with engineered, experimental animal models of disease. The integration of the QSP paradigm with the use of human MPS has the potential to enhance the process of drug discovery and development.
Collapse
Affiliation(s)
- D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark E Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chakra S Chennubhotla
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Lefever
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Fen Pei
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Faeder
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy R Lezon
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Del Favero G, Zaharescu R, Marko D. Functional impairment triggered by altertoxin II (ATXII) in intestinal cells in vitro: cross-talk between cytotoxicity and mechanotransduction. Arch Toxicol 2018; 92:3535-3547. [PMID: 30276433 PMCID: PMC6290659 DOI: 10.1007/s00204-018-2317-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022]
Abstract
Intestinal cells are able to continuously integrate response to multiple stimuli/stressors; these include the concomitant activation of “chemically driven” pathways, of paramount importance in the response to toxicants, as well as physical stimulation derived from motility. Altertoxin II (ATXII, 0.1, 1 and 10 µM), a mycotoxin produced by the food contaminant fungus Alternaria alternata was studied in HT-29 intestinal adenocarcinoma cells and in non-transformed intestinal epithelial cells, HCEC. One-hour incubation with ATXII was sufficient to trigger irreversible cytotoxicity in both cell types, as well as to modify cellular responses to concomitant pro-oxidant challenge (H2O2, 100–500 µM, DCF-DA assay) suggesting that even relatively short-time exposure of the intestinal cells could be sufficient to alter their functionality. Combination of ATXII (1 µM) with physical stimulation typical of the intestinal compartment (shear stress) revealed differential response of tumor-derived epithelial cells HT-29 in comparison to HCEC, in particular in the localization of the transcription factor Nrf2 (NF-E2-related factor 2). Moreover, ATXII reduced the migratory potential of HCEC as well as their membrane fluidity, but had no respective impact on HT-29 cells. Taken together, ATXII appeared to alter predominantly membrane functionality in HCEC thus hampering crucial functions for cellular motility/turnover, as well as barrier function of healthy intestinal cells and had very limited activity on the tumor counterparts.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090, Vienna, Austria.
| | - Ronita Zaharescu
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090, Vienna, Austria
| |
Collapse
|
24
|
Barbé L, Le Moullac-Vaidye B, Echasserieau K, Bernardeau K, Carton T, Bovin N, Nordgren J, Svensson L, Ruvoën-Clouet N, Le Pendu J. Histo-blood group antigen-binding specificities of human rotaviruses are associated with gastroenteritis but not with in vitro infection. Sci Rep 2018; 8:12961. [PMID: 30154494 PMCID: PMC6113245 DOI: 10.1038/s41598-018-31005-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/10/2018] [Indexed: 12/30/2022] Open
Abstract
Human strains of rotavirus A (RVAs) recognize fucosylated glycans belonging to histo-blood group antigens (HBGAs) through their spike protein VP8*. Lack of these ligands due to genetic polymorphisms is associated with resistance to gastroenteritis caused by P[8] genotype RVAs. With the aim to delineate the contribution of HBGAs in the process, we analyzed the glycan specificity of VP8* proteins from various P genotypes. Binding to saliva of VP8* from P[8] and P[4] genotypes required expression of both FUT2 and FUT3 enzymes, whilst binding of VP8* from the P[14] genotype required FUT2 and A enzymes. We further defined a glycan motif, GlcNAcβ3Galβ4GlcNAc, recognized by P[6] clinical strains. Conversion into Lewis antigens by the FUT3 enzyme impaired recognition, explaining their lower binding to saliva of Lewis positive phenotype. In addition, the presence of neutralizing antibodies was associated with the presence of the FUT2 wild type allele in sera from young healthy adults. Nonetheless, in vitro infection of transformed cell lines was independent of HBGAs expression, indicating that HBGAs are not human RV receptors. The match between results from saliva-based binding assays and the epidemiological data indicates that the polymorphism of human HBGAs controls susceptibility to RVAs, although the exact mechanism remains unclear.
Collapse
Affiliation(s)
- Laure Barbé
- CRCINA, Inserm, Université d'Angers, Université de Nantes, Nantes, France
| | | | - Klara Echasserieau
- CRCINA, Inserm, Université d'Angers, Université de Nantes, Nantes, France
- Plateforme P2R « Production de protéines recombinantes », SFR Sante F. Bonamy-IRS-UN, Université de Nantes, INSERM, CNRS, CHU Nantes, Nantes, France
| | - Karine Bernardeau
- CRCINA, Inserm, Université d'Angers, Université de Nantes, Nantes, France
- Plateforme P2R « Production de protéines recombinantes », SFR Sante F. Bonamy-IRS-UN, Université de Nantes, INSERM, CNRS, CHU Nantes, Nantes, France
| | | | - Nicolai Bovin
- Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Johan Nordgren
- Division of Molecular Virology, Medical Faculty, University of Linköping, Linköping, Sweden
| | - Lennart Svensson
- Division of Molecular Virology, Medical Faculty, University of Linköping, Linköping, Sweden
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Nathalie Ruvoën-Clouet
- CRCINA, Inserm, Université d'Angers, Université de Nantes, Nantes, France
- Oniris, Ecole Nationale Vétérinaire, Agroalimentaire et de l'Alimentation, Nantes, France
| | - Jacques Le Pendu
- CRCINA, Inserm, Université d'Angers, Université de Nantes, Nantes, France.
| |
Collapse
|
25
|
Low LA, Tagle DA. ‘You-on-a-chip’ for precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1456333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Lucie A. Low
- National Center for Advancing Translational Sciences, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Danilo A. Tagle
- National Center for Advancing Translational Sciences, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Watson DE, Hunziker R, Wikswo JP. Fitting tissue chips and microphysiological systems into the grand scheme of medicine, biology, pharmacology, and toxicology. Exp Biol Med (Maywood) 2017; 242:1559-1572. [PMID: 29065799 DOI: 10.1177/1535370217732765] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microphysiological systems (MPS), which include engineered organoids (EOs), single organ/tissue chips (TCs), and multiple organs interconnected to create miniature in vitro models of human physiological systems, are rapidly becoming effective tools for drug development and the mechanistic understanding of tissue physiology and pathophysiology. The second MPS thematic issue of Experimental Biology and Medicine comprises 15 articles by scientists and engineers from the National Institutes of Health, the IQ Consortium, the Food and Drug Administration, and Environmental Protection Agency, an MPS company, and academia. Topics include the progress, challenges, and future of organs-on-chips, dissemination of TCs into Pharma, children's health protection, liver zonation, liver chips and their coupling to interconnected systems, gastrointestinal MPS, maturation of immature cardiomyocytes in a heart-on-a-chip, coculture of multiple cell types in a human skin construct, use of synthetic hydrogels to create EOs that form neural tissue models, the blood-brain barrier-on-a-chip, MPS models of coupled female reproductive organs, coupling MPS devices to create a body-on-a-chip, and the use of a microformulator to recapitulate endocrine circadian rhythms. While MPS hardware has been relatively stable since the last MPS thematic issue, there have been significant advances in cell sourcing, with increased reliance on human-induced pluripotent stem cells, and in characterization of the genetic and functional cell state in MPS bioreactors. There is growing appreciation of the need to minimize perfusate-to-cell-volume ratios and respect physiological scaling of coupled TCs. Questions asked by drug developers are followed by an analysis of the potential value, costs, and needs of Pharma. Of highest value and lowest switching costs may be the development of MPS disease models to aid in the discovery of disease mechanisms; novel compounds including probes, leads, and clinical candidates; and mechanism of action of drug candidates. Impact statement Microphysiological systems (MPS), which include engineered organoids and both individual and coupled organs-on-chips and tissue chips, are a rapidly growing topic of research that addresses the known limitations of conventional cellular monoculture on flat plastic - a well-perfected set of techniques that produces reliable, statistically significant results that may not adequately represent human biology and disease. As reviewed in this article and the others in this thematic issue, MPS research has made notable progress in the past three years in both cell sourcing and characterization. As the field matures, currently identified challenges are being addressed, and new ones are being recognized. Building upon investments by the Defense Advanced Research Projects Agency, National Institutes of Health, Food and Drug Administration, Defense Threat Reduction Agency, and Environmental Protection Agency of more than $200 million since 2012 and sizable corporate spending, academic and commercial players in the MPS community are demonstrating their ability to meet the translational challenges required to apply MPS technologies to accelerate drug development and advance toxicology.
Collapse
Affiliation(s)
| | - Rosemarie Hunziker
- 2 National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Wikswo
- 3 Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics & Astronomy, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1807, USA
| |
Collapse
|