1
|
Ryoo H, Kimmel H, Rondo E, Underhill GH. Advances in high throughput cell culture technologies for therapeutic screening and biological discovery applications. Bioeng Transl Med 2024; 9:e10627. [PMID: 38818120 PMCID: PMC11135158 DOI: 10.1002/btm2.10627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 06/01/2024] Open
Abstract
Cellular phenotypes and functional responses are modulated by the signals present in their microenvironment, including extracellular matrix (ECM) proteins, tissue mechanical properties, soluble signals and nutrients, and cell-cell interactions. To better recapitulate and analyze these complex signals within the framework of more physiologically relevant culture models, high throughput culture platforms can be transformative. High throughput methodologies enable scientists to extract increasingly robust and broad datasets from individual experiments, screen large numbers of conditions for potential hits, better qualify and predict responses for preclinical applications, and reduce reliance on animal studies. High throughput cell culture systems require uniformity, assay miniaturization, specific target identification, and process simplification. In this review, we detail the various techniques that researchers have used to face these challenges and explore cellular responses in a high throughput manner. We highlight several common approaches including two-dimensional multiwell microplates, microarrays, and microfluidic cell culture systems as well as unencapsulated and encapsulated three-dimensional high throughput cell culture systems, featuring multiwell microplates, micromolds, microwells, microarrays, granular hydrogels, and cell-encapsulated microgels. We also discuss current applications of these high throughput technologies, namely stem cell sourcing, drug discovery and predictive toxicology, and personalized medicine, along with emerging opportunities and future impact areas.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Hannah Kimmel
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Evi Rondo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Gregory H. Underhill
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
2
|
Pike CM, Zwarycz B, McQueen BE, Castillo M, Barron C, Morowitz JM, Levi JA, Phadke D, Balik-Meisner M, Mav D, Shah R, Glasspoole DLC, Laetham R, Thelin W, Bunger MK, Boazak EM. Characterization and optimization of variability in a human colonic epithelium culture model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.559007. [PMID: 37790345 PMCID: PMC10542543 DOI: 10.1101/2023.09.22.559007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Animal models have historically been poor preclinical predictors of gastrointestinal (GI) directed therapeutic efficacy and drug-induced GI toxicity. Human stem and primary cell-derived culture systems are a major focus of efforts to create biologically relevant models that enhance preclinical predictive value of intestinal efficacy and toxicity. The inherent variability in stem-cell-based complex cultures makes development of useful models a challenge; the stochastic nature of stem-cell differentiation interferes with the ability to build and validate robust, reproducible assays that query drug responses and pharmacokinetics. In this study, we aimed to characterize and reduce potential sources of variability in a complex stem cell-derived intestinal epithelium model, termed RepliGut® Planar, across cells from multiple human donors, cell lots, and passage numbers. Assessment criteria included barrier formation and integrity, gene expression, and cytokine responses. Gene expression and culture metric analyses revealed that controlling for stem/progenitor-cell passage number reduces variability and maximizes physiological relevance of the model. After optimizing passage number, donor-specific differences in cytokine responses were observed in a case study, suggesting biologic variability is observable in cell cultures derived from multiple human sources. Our findings highlight key considerations for designing assays that can be applied to additional primary-cell derived systems, as well as establish utility of the RepliGut® Planar platform for robust development of human-predictive drug-response assays.
Collapse
|
3
|
Sockell A, Wong W, Longwell S, Vu T, Karlsson K, Mokhtari D, Schaepe J, Lo YH, Cornelius V, Kuo C, Van Valen D, Curtis C, Fordyce PM. A microwell platform for high-throughput longitudinal phenotyping and selective retrieval of organoids. Cell Syst 2023; 14:764-776.e6. [PMID: 37734323 DOI: 10.1016/j.cels.2023.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/24/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023]
Abstract
Organoids are powerful experimental models for studying the ontogeny and progression of various diseases including cancer. Organoids are conventionally cultured in bulk using an extracellular matrix mimic. However, bulk-cultured organoids physically overlap, making it impossible to track the growth of individual organoids over time in high throughput. Moreover, local spatial variations in bulk matrix properties make it difficult to assess whether observed phenotypic heterogeneity between organoids results from intrinsic cell differences or differences in the microenvironment. Here, we developed a microwell-based method that enables high-throughput quantification of image-based parameters for organoids grown from single cells, which can further be retrieved from their microwells for molecular profiling. Coupled with a deep learning image-processing pipeline, we characterized phenotypic traits including growth rates, cellular movement, and apical-basal polarity in two CRISPR-engineered human gastric organoid models, identifying genomic changes associated with increased growth rate and changes in accessibility and expression correlated with apical-basal polarity. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Alexandra Sockell
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Wing Wong
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Scott Longwell
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Thy Vu
- Department of Biochemistry, UT Austin, Austin, TX 78712, USA
| | - Kasper Karlsson
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel Mokhtari
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA
| | - Julia Schaepe
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yuan-Hung Lo
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Vincent Cornelius
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Calvin Kuo
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David Van Valen
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Christina Curtis
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94110, USA.
| | - Polly M Fordyce
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94110, USA; ChEM-H Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Luo Y, Li X, Zhao Y, Zhong W, Xing M, Lyu G. Development of Organs-on-Chips and Their Impact on Precision Medicine and Advanced System Simulation. Pharmaceutics 2023; 15:2094. [PMID: 37631308 PMCID: PMC10460056 DOI: 10.3390/pharmaceutics15082094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Drugs may undergo costly preclinical studies but still fail to demonstrate their efficacy in clinical trials, which makes it challenging to discover new drugs. Both in vitro and in vivo models are essential for disease research and therapeutic development. However, these models cannot simulate the physiological and pathological environment in the human body, resulting in limited drug detection and inaccurate disease modelling, failing to provide valid guidance for clinical application. Organs-on-chips (OCs) are devices that serve as a micro-physiological system or a tissue-on-a-chip; they provide accurate insights into certain functions and the pathophysiology of organs to precisely predict the safety and efficiency of drugs in the body. OCs are faster, more economical, and more precise. Thus, they are projected to become a crucial addition to, and a long-term replacement for, traditional preclinical cell cultures, animal studies, and even human clinical trials. This paper first outlines the nature of OCs and their significance, and then details their manufacturing-related materials and methodology. It also discusses applications of OCs in drug screening and disease modelling and treatment, and presents the future perspective of OCs.
Collapse
Affiliation(s)
- Ying Luo
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (Y.L.); (X.L.)
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xiaoxiao Li
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (Y.L.); (X.L.)
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
- Department of General Surgery, Huai’an 82 Hospital, Huai’an 223003, China
| | - Yawei Zhao
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (Y.Z.); (W.Z.)
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Wen Zhong
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (Y.Z.); (W.Z.)
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Guozhong Lyu
- Burn & Trauma Treatment Center, The Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (Y.L.); (X.L.)
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi 214000, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
- National Research Center for Emergency Medicine, Beijing 100000, China
| |
Collapse
|
5
|
Guevara-Garcia A, Soleilhac M, Minc N, Delacour D. Regulation and functions of cell division in the intestinal tissue. Semin Cell Dev Biol 2023:S1084-9521(23)00004-6. [PMID: 36702722 DOI: 10.1016/j.semcdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/16/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023]
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing epithelial tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs to ensure correct organ formation and functioning. In these processes, proliferation rates and division orientation regulate the speed, timing and direction of tissue expansion but also its proper patterning. Moreover, tissue homeostasis relies on spatio-temporal modulations of daughter cell behavior and arrangement. These aspects are particularly crucial in the intestine, which is one of the most proliferative tissues in adults, making it a very attractive adult organ system to study the role of cell division on epithelial morphogenesis and organ function. Although epithelial cell division has been the subject of intense research for many years in multiple models, it still remains in its infancy in the context of the intestinal tissue. In this review, we focus on the current knowledge on cell division and regulatory mechanisms at play in the intestinal epithelial tissue, as well as their importance in developmental biology and physiopathology.
Collapse
Affiliation(s)
| | - Matis Soleilhac
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Nicolas Minc
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Delphine Delacour
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France.
| |
Collapse
|
6
|
Zhang D, Qiao L. Intestine‐on‐a‐chip for intestinal disease study and pharmacological research. VIEW 2022. [DOI: 10.1002/viw.20220037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Dongxue Zhang
- Department of Chemistry, Institutes of Biomedical Sciences, and Shanghai Stomatological Hospital Fudan University Shanghai China
| | - Liang Qiao
- Department of Chemistry, Institutes of Biomedical Sciences, and Shanghai Stomatological Hospital Fudan University Shanghai China
| |
Collapse
|
7
|
Sullivan KM, Ko E, Kim EM, Ballance WC, Ito JD, Chalifoux M, Kim YJ, Bashir R, Kong H. Extracellular Microenvironmental Control for Organoid Assembly. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1209-1222. [PMID: 35451330 PMCID: PMC9836674 DOI: 10.1089/ten.teb.2021.0186] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/04/2022] [Indexed: 01/22/2023]
Abstract
Organoids, which are multicellular clusters with similar physiological functions to living organs, have gained increasing attention in bioengineering. As organoids become more advanced, methods to form complex structures continue to develop. There is evidence that the extracellular microenvironment can regulate organoid quality. The extracellular microenvironment consists of soluble bioactive molecules, extracellular matrix, and biofluid flow. However, few efforts have been made to discuss the microenvironment optimal to engineer specific organoids. Therefore, this review article examines the extent to which engineered extracellular microenvironments regulate organoid quality. First, we summarize the natural tissue and organ's unique chemical and mechanical properties, guiding researchers to design an extracellular microenvironment used for organoid engineering. Then, we summarize how the microenvironments contribute to the formation and growth of the brain, lung, intestine, liver, retinal, and kidney organoids. The approaches to forming and evaluating the resulting organoids are also discussed in detail. Impact statement Organoids, which are multicellular clusters with similar physiological function to living organs, have been gaining increasing attention in bioengineering. As organoids become more advanced, methods to form complex structures continue to develop. This review article focuses on recent efforts to engineer the extracellular microenvironment in organoid research. We summarized the natural organ's microenvironment, which informs researchers of key factors that can influence organoid formation. Then, we summarize how these microenvironmental controls significantly contribute to the formation and growth of the corresponding brain, lung, intestine, liver, retinal, and kidney organoids. The approaches to forming and evaluating the resulting organoids are discussed in detail, including extracellular matrix choice and properties, culture methods, and the evaluation of the morphology and functionality through imaging and biochemical analysis.
Collapse
Affiliation(s)
- Kathryn M. Sullivan
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Eunkyung Ko
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Eun Mi Kim
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - William C. Ballance
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - John D. Ito
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Madeleine Chalifoux
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST–Europe), Saarbrucken, Germany
| | - Rashid Bashir
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| |
Collapse
|
8
|
Current Knowledge About the Implication of Bacterial Microbiota in Human Health and Disease. ACTA MEDICA BULGARICA 2021. [DOI: 10.2478/amb-2021-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Recent advances in molecular genetics and the invention of new technologies led to a development in our knowledge about human microbiota, specifically bacterial one. The microbiota plays a fundamental role in the immunologic, hormonal and metabolic homeostasis of the host. After the initiation of the Human Microbiome Project, it became clear that the human microbiota consists of the 10-100 trillion symbiotic microbial cells harbored by each person, primarily bacteria in the gut, but also in other spots as the skin, mouth, nose, and vagina. Despite of the differences in studying bacterial species, decreased bacterial diversity and persistence has been connected with several diverse human diseases primarily diabetes, IBD (inflammatory bowel disease) and others; attempts were made even to explain psychiatric pathology. Several species emerged as dominant and were clearly linked to certain disorders or accepted as biomarkers of others. The current review aims to discuss key issues of our current knowledge about bacteria in human, the difficulties and methods of its analysis, its contribution to human health and responsibility for human diseases.
Collapse
|
9
|
Boeri L, Perottoni S, Izzo L, Giordano C, Albani D. Microbiota-Host Immunity Communication in Neurodegenerative Disorders: Bioengineering Challenges for In Vitro Modeling. Adv Healthc Mater 2021; 10:e2002043. [PMID: 33661580 PMCID: PMC11468246 DOI: 10.1002/adhm.202002043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Human microbiota communicates with its host by secreting signaling metabolites, enzymes, or structural components. Its homeostasis strongly influences the modulation of human tissue barriers and immune system. Dysbiosis-induced peripheral immunity response can propagate bacterial and pro-inflammatory signals to the whole body, including the brain. This immune-mediated communication may contribute to several neurodegenerative disorders, as Alzheimer's disease. In fact, neurodegeneration is associated with dysbiosis and neuroinflammation. The interplay between the microbial communities and the brain is complex and bidirectional, and a great deal of interest is emerging to define the exact mechanisms. This review focuses on microbiota-immunity-central nervous system (CNS) communication and shows how gut and oral microbiota populations trigger immune cells, propagating inflammation from the periphery to the cerebral parenchyma, thus contributing to the onset and progression of neurodegeneration. Moreover, an overview of the technological challenges with in vitro modeling of the microbiota-immunity-CNS axis, offering interesting technological hints about the most advanced solutions and current technologies is provided.
Collapse
Affiliation(s)
- Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Diego Albani
- Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSvia Mario Negri 2Milan20156Italy
| |
Collapse
|
10
|
Sung JH. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin Drug Metab Toxicol 2021; 17:969-986. [PMID: 33764248 DOI: 10.1080/17425255.2021.1908996] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Accurate prediction of pharmacokinetic (PK) and toxicokinetics (TK) of drugs is imperative for successful development of new pharmaceutics. Although conventional in vitro methods for predicting the PK and TK of drugs are well established, limitations still exist and more advanced chip-based in vitro platforms combined with mathematical models can help researchers overcome the limitations. Areas covered: We will review recent progress in the development of multi-organ-on-a-chip platforms for predicting PK and TK of drugs, as well as mathematical approaches that can be combined with these platforms for experiment design, data analysis and in vitro-in vivo extrapolation (IVIVE) for application to humans. Expert opinion: Although there remain some challenges to be addressed, the remarkable progress in the area of multi-organ-on-a-chip in recent years indicate that we will see tangible outcomes that can be utilized in the pharmaceutical industry in near future.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, sejong, Republic of Korea
| |
Collapse
|
11
|
Arnold JW, Roach J, Fabela S, Moorfield E, Ding S, Blue E, Dagher S, Magness S, Tamayo R, Bruno-Barcena JM, Azcarate-Peril MA. The pleiotropic effects of prebiotic galacto-oligosaccharides on the aging gut. MICROBIOME 2021; 9:31. [PMID: 33509277 PMCID: PMC7845053 DOI: 10.1186/s40168-020-00980-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/16/2020] [Indexed: 05/05/2023]
Abstract
BACKGROUND Prebiotic galacto-oligosaccharides (GOS) have an extensively demonstrated beneficial impact on intestinal health. In this study, we determined the impact of GOS diets on hallmarks of gut aging: microbiome dysbiosis, inflammation, and intestinal barrier defects ("leaky gut"). We also evaluated if short-term GOS feeding influenced how the aging gut responded to antibiotic challenges in a mouse model of Clostridioides difficile infection. Finally, we assessed if colonic organoids could reproduce the GOS responder-non-responder phenotypes observed in vivo. RESULTS Old animals had a distinct microbiome characterized by increased ratios of non-saccharolytic versus saccharolytic bacteria and, correspondingly, a lower abundance of β-galactosidases compared to young animals. GOS reduced the overall diversity, increased the abundance of specific saccharolytic bacteria (species of Bacteroides and Lactobacillus), increased the abundance of β-galactosidases in young and old animals, and increased the non-saccharolytic organisms; however, a robust, homogeneous bifidogenic effect was not observed. GOS reduced age-associated increased intestinal permeability and increased MUC2 expression and mucus thickness in old mice. Clyndamicin reduced the abundance Bifidobacterium while increasing Akkermansia, Clostridium, Coprococcus, Bacillus, Bacteroides, and Ruminococcus in old mice. The antibiotics were more impactful than GOS on modulating serum markers of inflammation. Higher serum levels of IL-17 and IL-6 were observed in control and GOS diets in the antibiotic groups, and within those groups, levels of IL-6 were higher in the GOS groups, regardless of age, and higher in the old compared to young animals in the control diet groups. RTqPCR revealed significantly increased gene expression of TNFα in distal colon tissue of old mice, which was decreased by the GOS diet. Colon transcriptomics analysis of mice fed GOS showed increased expression of genes involved in small-molecule metabolic processes and specifically the respirasome in old animals, which could indicate an increased oxidative metabolism and energetic efficiency. In young mice, GOS induced the expression of binding-related genes. The galectin gene Lgals1, a β-galactosyl-binding lectin that bridges molecules by their sugar moieties and is an important modulator of the immune response, and the PI3K-Akt and ECM-receptor interaction pathways were also induced in young mice. Stools from mice exhibiting variable bifidogenic response to GOS injected into colon organoids in the presence of prebiotics reproduced the response and non-response phenotypes observed in vivo suggesting that the composition and functionality of the microbiota are the main contributors to the phenotype. CONCLUSIONS Dietary GOS modulated homeostasis of the aging gut by promoting changes in microbiome composition and host gene expression, which was translated into decreased intestinal permeability and increased mucus production. Age was a determining factor on how prebiotics impacted the microbiome and expression of intestinal epithelial cells, especially apparent from the induction of galectin-1 in young but not old mice. Video abstract.
Collapse
Affiliation(s)
- Jason W Arnold
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffery Roach
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- UNC Information Technology Services and Research Computing, University of North Carolina, Chapel Hill, NC, USA
| | - Salvador Fabela
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Current affiliation: Programa de Inmunología Molecular Microbiana. Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Emily Moorfield
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Shengli Ding
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Eric Blue
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Suzanne Dagher
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Scott Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Rita Tamayo
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Jose M Bruno-Barcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Kang SM, Kim D, Lee JH, Takayama S, Park JY. Engineered Microsystems for Spheroid and Organoid Studies. Adv Healthc Mater 2021; 10:e2001284. [PMID: 33185040 PMCID: PMC7855453 DOI: 10.1002/adhm.202001284] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/01/2020] [Indexed: 01/09/2023]
Abstract
3D in vitro model systems such as spheroids and organoids provide an opportunity to extend the physiological understanding using recapitulated tissues that mimic physiological characteristics of in vivo microenvironments. Unlike 2D systems, 3D in vitro systems can bridge the gap between inadequate 2D cultures and the in vivo environments, providing novel insights on complex physiological mechanisms at various scales of organization, ranging from the cellular, tissue-, to organ-levels. To satisfy the ever-increasing need for highly complex and sophisticated systems, many 3D in vitro models with advanced microengineering techniques have been developed to answer diverse physiological questions. This review summarizes recent advances in engineered microsystems for the development of 3D in vitro model systems. The relationship between the underlying physics behind the microengineering techniques, and their ability to recapitulate distinct 3D cellular structures and functions of diverse types of tissues and organs are highlighted and discussed in detail. A number of 3D in vitro models and their engineering principles are also introduced. Finally, current limitations are summarized, and perspectives for future directions in guiding the development of 3D in vitro model systems using microengineering techniques are provided.
Collapse
Affiliation(s)
- Sung-Min Kang
- Department of Green Chemical Engineering, Sangmyung University, Cheonan, Chungnam, 31066, Republic of Korea
| | - Daehan Kim
- Department of Mechanical Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji-Hoon Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Joong Yull Park
- Department of Mechanical Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
13
|
Jia Z, Wang Y, Cao L, Wang Y, Song Y, Yang X, Bing Z, Cao Z, Liu P, Zhang S, Chen Z, Huang M, Yu Y, Han-Zhang H, Song J, Christoph DC, Passaro A, Gridelli C, Hishida T, Liang N, Li S. First-line treatment selection with organoids of an EGFRm + TP53m stage IA1 patient with early metastatic recurrence after radical surgery and follow-up. J Thorac Dis 2020; 12:3764-3773. [PMID: 32802455 PMCID: PMC7399434 DOI: 10.21037/jtd-20-1882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ziqi Jia
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanyu Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Song
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoying Yang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhili Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Peng Liu
- Medical Research Center, Central Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuyang Zhang
- Medical Research Center, Central Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zexin Chen
- Accurate International Biotechnology Co. Ltd., Guangzhou, China
| | - Min Huang
- Accurate International Biotechnology Co. Ltd., Guangzhou, China
| | - Yan Yu
- Accurate International Biotechnology Co. Ltd., Guangzhou, China
| | - Han Han-Zhang
- Burning Rock Biotechnology Co. Ltd., Guangzhou, China
| | - Jinlei Song
- Burning Rock Biotechnology Co. Ltd., Guangzhou, China
| | - Daniel C Christoph
- Department of Medical Oncology/Haematology, Evang. Kliniken Essen-Mitte, Essen, Germany
| | - Antonio Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, "S.G. Moscati" Hospital, Contrada Amoretta, 83100, Avellino, Italy
| | - Tomoyuki Hishida
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Shin W, Ambrosini YM, Shin YC, Wu A, Min S, Koh D, Park S, Kim S, Koh H, Kim HJ. Robust Formation of an Epithelial Layer of Human Intestinal Organoids in a Polydimethylsiloxane-Based Gut-on-a-Chip Microdevice. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2. [PMID: 33532747 PMCID: PMC7849371 DOI: 10.3389/fmedt.2020.00002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polydimethylsiloxane (PDMS) is a silicone polymer that has been predominantly used in a human organ-on-a-chip microphysiological system. The hydrophobic surface of a microfluidic channel made of PDMS often results in poor adhesion of the extracellular matrix (ECM) as well as cell attachment. The surface modification by plasma or UV/ozone treatment in a PDMS-based device produces a hydrophilic surface that allows robust ECM coating and the reproducible attachment of human intestinal immortalized cell lines. However, these surface-activating methods have not been successful in forming a monolayer of the biopsy-derived primary organoid epithelium. Several existing protocols to grow human intestinal organoid cells in a PDMS microchannel are not always reproducibly operative due to the limited information. Here, we report an optimized methodology that enables robust and reproducible attachment of the intestinal organoid epithelium in a PDMS-based gut-on-a-chip. Among several reported protocols, we optimized a method by performing polyethyleneimine-based surface functionalization followed by the glutaraldehyde cross linking to activate the PDMS surface. Moreover, we discovered that the post-functionalization step contributes to provide uniform ECM deposition that allows to produce a robust attachment of the dissociated intestinal organoid epithelium in a PDMS-based microdevice. We envision that our optimized protocol may disseminate an enabling methodology to advance the integration of human organotypic cultures in a human organ-on-a-chip for patient-specific disease modeling.
Collapse
Affiliation(s)
- Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Yoko M Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Alexander Wu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Domin Koh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Sowon Park
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Kim
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Hong Koh
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
15
|
Hewes SA, Wilson RL, Estes MK, Shroyer NF, Blutt SE, Grande-Allen KJ. In Vitro Models of the Small Intestine: Engineering Challenges and Engineering Solutions. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:313-326. [PMID: 32046599 PMCID: PMC7462033 DOI: 10.1089/ten.teb.2019.0334] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Pathologies affecting the small intestine contribute significantly to the disease burden of both the developing and the developed world, which has motivated investigation into the disease mechanisms through in vitro models. Although existing in vitro models recapitulate selected features of the intestine, various important aspects have often been isolated or omitted due to the anatomical and physiological complexity. The small intestine's intricate microanatomy, heterogeneous cell populations, steep oxygen gradients, microbiota, and intestinal wall contractions are often not included in in vitro experimental models of the small intestine, despite their importance in both intestinal biology and pathology. Known and unknown interdependencies between various physiological aspects necessitate more complex in vitro models. Microfluidic technology has made it possible to mimic the dynamic mechanical environment, signaling gradients, and other important aspects of small intestinal biology. This review presents an overview of the complexity of small intestinal anatomy and bioengineered models that recapitulate some of these physiological aspects.
Collapse
Affiliation(s)
- Sarah A. Hewes
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Reid L. Wilson
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
16
|
Guenat OT, Geiser T, Berthiaume F. Clinically Relevant Tissue Scale Responses as New Readouts from Organs-on-a-Chip for Precision Medicine. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:111-133. [PMID: 31961712 DOI: 10.1146/annurev-anchem-061318-114919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Organs-on-chips (OOC) are widely seen as being the next generation in vitro models able to accurately recreate the biochemical-physical cues of the cellular microenvironment found in vivo. In addition, they make it possible to examine tissue-scale functional properties of multicellular systems dynamically and in a highly controlled manner. Here we summarize some of the most remarkable examples of OOC technology's ability to extract clinically relevant tissue-level information. The review is organized around the types of OOC outputs that can be measured from the cultured tissues and transferred to clinically meaningful information. First, the creation of functional tissues-on-chip is discussed, followed by the presentation of tissue-level readouts specific to OOC, such as morphological changes, vessel formation and function, tissue properties, and metabolic functions. In each case, the clinical relevance of the extracted information is highlighted.
Collapse
Affiliation(s)
- Olivier T Guenat
- ARTORG Center for Biomedical Engineering Research, Medical Faculty, University of Bern, CH-3008 Bern, Switzerland;
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
- Thoracic Surgery Department, University Hospital of Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
17
|
Ambrosini YM, Shin W, Min S, Kim HJ. Microphysiological Engineering of Immune Responses in Intestinal Inflammation. Immune Netw 2020; 20:e13. [PMID: 32395365 PMCID: PMC7192834 DOI: 10.4110/in.2020.20.e13] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/18/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
The epithelial barrier in the gastrointestinal (GI) tract is a protective interface that endures constant exposure to the external environment while maintaining its close contact with the local immune system. Growing evidence has suggested that the intercellular crosstalk in the GI tract contributes to maintaining the homeostasis in coordination with the intestinal microbiome as well as the tissue-specific local immune elements. Thus, it is critical to map the complex crosstalks in the intestinal epithelial-microbiome-immune (EMI) axis to identify a pathological trigger in the development of intestinal inflammation, including inflammatory bowel disease. However, deciphering a specific contributor to the onset of pathophysiological cascades has been considerably hindered by the challenges in current in vivo and in vitro models. Here, we introduce various microphysiological engineering models of human immune responses in the EMI axis under the healthy conditions and gut inflammation. As a prospective model, we highlight how the human “gut inflammation-on-a-chip” can reconstitute the pathophysiological immune responses and contribute to understanding the independent role of inflammatory factors in the EMI axis on the initiation of immune responses under barrier dysfunction. We envision that the microengineered immune models can be useful to build a customizable patient's chip for the advance in precision medicine.
Collapse
Affiliation(s)
- Yoko M Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA.,Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011 USA
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Soyoun Min
- 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.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
18
|
Lee SH, Choi N, Sung JH. Pharmacokinetic and pharmacodynamic insights from microfluidic intestine-on-a-chip models. Expert Opin Drug Metab Toxicol 2019; 15:1005-1019. [PMID: 31794278 DOI: 10.1080/17425255.2019.1700950] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: After administration, a drug undergoes absorption, distribution, metabolism, and elimination (ADME) before exerting its effect on the body. The combination of these process yields the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of a drug. Although accurate prediction of PK and PD profiles is essential for drug development, conventional in vitro models are limited by their lack of physiological relevance. Recently, microtechnology-based in vitro model systems, termed 'organ-on-a-chip,' have emerged as a potential solution.Areas covered: Orally administered drugs are absorbed through the intestinal wall and transported to the liver before entering systemic circulation, which plays an important role in the PK and PD profiles. Recently developed, chip-based in vitro models can be useful models for simulating such processes and will be covered in this paper.Expert opinion: The potential of intestine-on-a-chip models combined with conventional PK-PD modeling has been demonstrated with promising preliminary results. However, there are several challenges to overcome. Development of the intestinal wall, integration of the gut microbiome, and the provision of an intestine-specific environment must be achieved to realize in vivo-like intestinal model and enhance the efficiency of drug development.
Collapse
Affiliation(s)
- Seung Hwan Lee
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Costa J, Ahluwalia A. Advances and Current Challenges in Intestinal in vitro Model Engineering: A Digest. Front Bioeng Biotechnol 2019; 7:144. [PMID: 31275931 PMCID: PMC6591368 DOI: 10.3389/fbioe.2019.00144] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022] Open
Abstract
The physiological environment of the intestine is characterized by its variegated composition, numerous functions and unique dynamic conditions, making it challenging to recreate the organ in vitro. This review outlines the requirements for engineering physiologically relevant intestinal in vitro models, mainly focusing on the importance of the mechano-structural cues that are often neglected in classic cell culture systems. More precisely: the topography, motility and flow present in the intestinal epithelium. After defining quantitative descriptors for these features, we describe the current state of the art, citing relevant approaches used to address one (or more) of the elements in question, pursuing a progressive conceptual construction of an "ideal" biomimetic intestinal model. The review concludes with a critical assessment of the currently available methods to summarize the important features of the intestinal tissue in the light of their different applications.
Collapse
Affiliation(s)
| | - Arti Ahluwalia
- Research Center “E. Piaggio” and Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
20
|
Zakhem E, Raghavan S, Suhar RA, Bitar KN. Bioengineering and regeneration of gastrointestinal tissue: where are we now and what comes next? Expert Opin Biol Ther 2019; 19:527-537. [PMID: 30880502 DOI: 10.1080/14712598.2019.1595579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The field of tissue engineering and regenerative medicine has been applied to the gastrointestinal (GI) tract for a couple decades. Several achievements have been accomplished that provide promising tools for treating diseases of the GI tract. AREAS COVERED The work described in this review covers the traditional aspect of using cells and scaffolds to replace parts of the tract. Several studies investigated different types of biomaterials and different types of cells. A more recent approach involved the use of gut-derived organoid units that can differentiate into all gut cell layers. The most recent approach introduced the use of organ-on-a-chip concept to understand the physiology and pathophysiology of the GI system. EXPERT OPINION The different approaches tackle the diseases of the GI tract from different perspectives. While all these different approaches provide a promising and encouraging future for this field, the translational aspect is yet to be studied.
Collapse
Affiliation(s)
- Elie Zakhem
- a Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine , Winston Salem , NC , USA.,b Section on Gastroenterology , Wake Forest School of Medicine , Winston Salem , NC , USA
| | - Shreya Raghavan
- c Department of Materials Science and Engineering , University of Michigan , Ann Arbor , MI , USA
| | - Riley A Suhar
- d Department of Materials Science and Engineering , Stanford University , Stanford , CA , USA
| | - Khalil N Bitar
- a Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine , Winston Salem , NC , USA.,b Section on Gastroenterology , Wake Forest School of Medicine , Winston Salem , NC , USA.,e Virginia Tech-Wake Forest School of Biomedical Engineering Sciences , Winston-Salem , NC , USA
| |
Collapse
|
21
|
Oksdath M, Perrin SL, Bardy C, Hilder EF, DeForest CA, Arrua RD, Gomez GA. Review: Synthetic scaffolds to control the biochemical, mechanical, and geometrical environment of stem cell-derived brain organoids. APL Bioeng 2018; 2:041501. [PMID: 31069322 PMCID: PMC6481728 DOI: 10.1063/1.5045124] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/31/2018] [Indexed: 01/16/2023] Open
Abstract
Stem cell-derived brain organoids provide a powerful platform for systematic studies of tissue functional architecture and the development of personalized therapies. Here, we review key advances at the interface of soft matter and stem cell biology on synthetic alternatives to extracellular matrices. We emphasize recent biomaterial-based strategies that have been proven advantageous towards optimizing organoid growth and controlling the geometrical, biomechanical, and biochemical properties of the organoid's three-dimensional environment. We highlight systems that have the potential to increase the translational value of region-specific brain organoid models suitable for different types of manipulations and high-throughput applications.
Collapse
Affiliation(s)
- Mariana Oksdath
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | - Sally L. Perrin
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | | | - Emily F. Hilder
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Cole A. DeForest
- Department of Chemical Engineering and Department of Bioengineering, University of Washington, Seattle, Washington 98195-1750, USA
| | - R. Dario Arrua
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Guillermo A. Gomez
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| |
Collapse
|
22
|
De Gregorio V, Imparato G, Urciuolo F, Netti PA. Micro-patterned endogenous stroma equivalent induces polarized crypt-villus architecture of human small intestinal epithelium. Acta Biomater 2018; 81:43-59. [PMID: 30282052 DOI: 10.1016/j.actbio.2018.09.061] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/29/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022]
Abstract
The small intestine is the major site for digestion, drug and nutrient absorption, as well as a primary site for many diseases. Current in vitro gut models fail in reproducing the complex intestinal extracellular matrix (ECM) network of the lamina propria and the peculiar architecture of the crypt-villus axis. Here we proposed a novel in vitro human intestine model that mimics the intestinal stromal topography and composition and strictly reproduces the tissue polarity with the crypt-villus architecture. First we developed a 3D human intestinal stromal equivalent (3D-ISE) composed of human intestinal subepithelial myofibroblasts (ISEMFs) embedded in their own extracellular matrix. Then, we seeded human colon carcinoma-derived cells (Caco-2) onto flat or patterned cell-synthetized stromal equivalent structure and cultured them until the formation of a well-oriented epithelium. We demonstrated that the patterned stroma increases the absorbing surface area, the epithelial proliferation rate, and the density of microvilli. In addition it induces changes in the biological functions of the epithelial cells such as enzymes and mucus production, polarization and tightness showing a physiological cell-lineage compartmentalization along the crypt/villi axes with the undifferentiated phenotypes at the base. At last, we reproduced an inflamed intestinal tissue model in which we identified the contribution of the stromal microenvironment by molecular (cytokines release and MMPs production) and immunofluorescence analyses and the effects of the epithelial-stromal cross-talk in the intestinal innate immunity by multiphoton investigation that revealed differences in the collagen network architecture. STATEMENT OF SIGNIFICANCE: The intestinal stroma morphology and composition has a fundamental role in crypt-villus development and appropriate epithelial cell-lineage compartmentalization. On this base, here we develop an engineered organotypic model of human intestine equivalent in which a functional epithelial/ECM crosstalk is recapitulated. Due to its accessible luminal surface it provides a new platform for preclinical studies of mucosal immunology and bowel inflammation as well as the assessment of pharmaco-toxicity studies.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy.
| | - Francesco Urciuolo
- Department of Chemical Materials and Industrial Production (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; Department of Chemical Materials and Industrial Production (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
23
|
Lee SY, Sung JH. Gut-liver on a chip toward an in vitro model of hepatic steatosis. Biotechnol Bioeng 2018; 115:2817-2827. [PMID: 29981260 DOI: 10.1002/bit.26793] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/31/2018] [Accepted: 07/02/2018] [Indexed: 12/16/2022]
Abstract
Hepatic steatosis is a process of abnormal lipid deposition within the liver cells, often caused by excessive alcohol uptake or obesity. A conventional in vitro model for hepatic steatosis uses a liver cell culture, treated with fatty acids and measures accumulation of lipids within the cells. This model does not recapitulate the complex process of absorption and metabolism of digestive lipids. Here, we introduce a gut-liver chip, which mimics the gut absorption and hepatic metabolism in a microfluidic chip. Absorption of fatty acids through gut layer and subsequent deposition within liver cells was demonstrated. Tumor necrosis factor-α, butyrate, and α-lipoic acid were chosen as model molecules that can affect hepatic steatosis via different mechanisms, and their effects were evaluated. Our results suggest that the gut-liver chip can mimic the absorption and accumulation of fatty acids in the gut and the liver.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Chemical Engineering, Hongik University, Seoul, Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, Korea
| |
Collapse
|
24
|
Parker A, Lawson MAE, Vaux L, Pin C. Host-microbe interaction in the gastrointestinal tract. Environ Microbiol 2018; 20:2337-2353. [PMID: 28892253 PMCID: PMC6175405 DOI: 10.1111/1462-2920.13926] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/25/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022]
Abstract
The gastrointestinal tract is a highly complex organ in which multiple dynamic physiological processes are tightly coordinated while interacting with a dense and extremely diverse microbial population. From establishment in early life, through to host-microbe symbiosis in adulthood, the gut microbiota plays a vital role in our development and health. The effect of the microbiota on gut development and physiology is highlighted by anatomical and functional changes in germ-free mice, affecting the gut epithelium, immune system and enteric nervous system. Microbial colonisation promotes competent innate and acquired mucosal immune systems, epithelial renewal, barrier integrity, and mucosal vascularisation and innervation. Interacting or shared signalling pathways across different physiological systems of the gut could explain how all these changes are coordinated during postnatal colonisation, or after the introduction of microbiota into germ-free models. The application of cell-based in-vitro experimental systems and mathematical modelling can shed light on the molecular and signalling pathways which regulate the development and maintenance of homeostasis in the gut and beyond.
Collapse
Affiliation(s)
- Aimée Parker
- Quadram Institute BioscienceNorwich Research ParkNR4 7UAUK
| | | | - Laura Vaux
- Quadram Institute BioscienceNorwich Research ParkNR4 7UAUK
| | - Carmen Pin
- Quadram Institute BioscienceNorwich Research ParkNR4 7UAUK
| |
Collapse
|
25
|
Zavrel EA, Shuler ML, Shen X. A SIMPLE ASPECT RATIO DEPENDENT METHOD OF PATTERNING MICROWELLS FOR SELECTIVE CELL ATTACHMENT. PROCEEDINGS OF THE ... DESIGN OF MEDICAL DEVICES CONFERENCE. DESIGN OF MEDICAL DEVICES CONFERENCE 2018; 2018:V001T05A001. [PMID: 30854173 PMCID: PMC6408146 DOI: 10.1115/dmd2018-6811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
3-D culture has been shown to provide cells with a more physiologically authentic environment than traditional 2-D (planar) culture [1, 2]. 3-D cues allow cells to exhibit more realistic functions and behaviors, e.g., adhesion, spreading, migration, metabolic activity, and differentiation. Knowledge of changes in cell morphology, mechanics, and mobility in response to geometrical cues and topological stimuli is important for understanding normal and pathological cell development [3]. Microfabrication provides unique in vitro approaches to recapitulating in vivo conditions due to the ability to precisely control the cellular microenvironment [4, 5]. Microwell arrays have emerged as robust alternatives to traditional 2D cell culture substrates as they are relatively simple and compatible with existing laboratory techniques and instrumentation [6, 7]. In particular, microwells have been adopted as a biomimetic approach to modeling the unique micro-architecture of the epithelial lining of the gastrointestinal (GI) tract [8–10]. The inner (lumen-facing) surface of the intestine has a convoluted topography consisting of finger-like projections (villi) with deep well-like invaginations (crypts) between them. The dimensions of villi and crypts are on the order of hundreds of microns (100–700 μm in height and 50–250 μm in diameter) [11]. While microwells have proven important in the development of physiologically realistic in vitro models of human intestine, existing methods of ensuring their surface is suitable for cell culture are lacking. Sometimes it is desirable to selectively seed cells within microwells and confine or restrict them to the microwells in which they are seeded. Existing methods of patterning microwells for cell attachment either lack selectivity, meaning cells can adhere and migrate anywhere on the microwell array, i.e., inside microwells or outside of them, or necessitate sophisticated techniques such as micro-contact printing, which requires precise alignment and control to selectively pattern the bottoms of microwells for cell attachment [12, 13].
Collapse
Affiliation(s)
- Erik A Zavrel
- Department of Biomedical Engineering, Cornell University Ithaca, NY, United States
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University Ithaca, NY, United States
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University Durham, NC, United States
| |
Collapse
|
26
|
Lee SH, Sung JH. Organ-on-a-Chip Technology for Reproducing Multiorgan Physiology. Adv Healthc Mater 2018; 7. [PMID: 28945001 DOI: 10.1002/adhm.201700419] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/04/2017] [Indexed: 12/14/2022]
Abstract
In the drug development process, the accurate prediction of drug efficacy and toxicity is important in order to reduce the cost, labor, and effort involved. For this purpose, conventional 2D cell culture models are used in the early phase of drug development. However, the differences between the in vitro and the in vivo systems have caused the failure of drugs in the later phase of the drug-development process. Therefore, there is a need for a novel in vitro model system that can provide accurate information for evaluating the drug efficacy and toxicity through a closer recapitulation of the in vivo system. Recently, the idea of using microtechnology for mimicking the microscale tissue environment has become widespread, leading to the development of "organ-on-a-chip." Furthermore, the system is further developed for realizing a multiorgan model for mimicking interactions between multiple organs. These advancements are still ongoing and are aimed at ultimately developing "body-on-a-chip" or "human-on-a-chip" devices for predicting the response of the whole body. This review summarizes recently developed organ-on-a-chip technologies, and their applications for reproducing multiorgan functions.
Collapse
Affiliation(s)
- Seung Hwan Lee
- School of Chemical and Biological Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering; Hongik University; Seoul 04066 Republic of Korea
| |
Collapse
|
27
|
Abstract
Current in vitro gut models lack physiological relevance, and various approaches have been taken to improve current cell culture models. For example, mimicking the three-dimensional (3D) tissue structure or fluidic environment has been shown to improve the physiological function of gut cells. Here, we incorporated a collagen scaffold that mimics the human intestinal villi into a microfluidic device, thus providing cells with both 3D tissue structure and fluidic shear. We hypothesized that the combined effect of 3D structure and fluidic shear may provide cells with adequate stimulus to induce further differentiation and improve physiological relevance. The physiological function of our '3D gut chip' was assessed by measuring the absorptive permeability of the gut epithelium and activity of representative enzymes, as well as morphological evaluation. Our results suggest that the combination of fluidic stimulus and 3D structure induces further improvement in gut functions. Our work provides insight into the effect of different tissue environment on gut cells.
Collapse
|
28
|
Wang Y, Kim R, Gunasekara DB, Reed MI, DiSalvo M, Nguyen DL, Bultman SJ, Sims CE, Magness ST, Allbritton NL. Formation of Human Colonic Crypt Array by Application of Chemical Gradients Across a Shaped Epithelial Monolayer. Cell Mol Gastroenterol Hepatol 2017; 5:113-130. [PMID: 29693040 PMCID: PMC5904049 DOI: 10.1016/j.jcmgh.2017.10.007] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS The successful culture of intestinal organoids has greatly enhanced our understanding of intestinal stem cell physiology and enabled the generation of novel intestinal disease models. Although of tremendous value, intestinal organoid culture systems have not yet fully recapitulated the anatomy or physiology of the in vivo intestinal epithelium. The aim of this work was to re-create an intestinal epithelium with a high density of polarized crypts that respond in a physiologic manner to addition of growth factors, metabolites, or cytokines to the basal or luminal tissue surface as occurs in vivo. METHODS A self-renewing monolayer of human intestinal epithelium was cultured on a collagen scaffold microfabricated with an array of crypt-like invaginations. Placement of chemical factors in either the fluid reservoir below or above the cell-covered scaffolding created a gradient of that chemical across the growing epithelial tissue possessing the in vitro crypt structures. Crypt polarization (size of the stem/proliferative and differentiated cell zones) was assessed in response to gradients of growth factors, cytokines, and bacterial metabolites. RESULTS Chemical gradients applied to the shaped human epithelium re-created the stem/proliferative and differentiated cell zones of the in vivo intestine. Short-chain fatty acids applied as a gradient from the luminal side confirmed long-standing hypotheses that butyrate diminished stem/progenitor cell proliferation and promoted differentiation into absorptive colonocytes. A gradient of interferon-γ and tumor necrosis factor-α significantly suppressed the stem/progenitor cell proliferation, altering crypt formation. CONCLUSIONS The in vitro human colon crypt array accurately mimicked the architecture, luminal accessibility, tissue polarity, cell migration, and cellular responses of in vivo intestinal crypts.
Collapse
Key Words
- ALP, alkaline phosphatase
- BSA, bovine serum albumin
- DM, differentiation medium
- DM-B, differentiation medium plus 5 mmol/L butyrate
- DM-D, DM plus 10 μmol/L DAPT
- EDC, 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride
- ELISA, enzyme-linked immunosorbent assay
- EM, expansion medium
- EdU, 5-ethynyl-20-deoxyuridine
- IFN-γ, interferon-γ
- Intestinal Epithelial Cells
- Intestine-On-A-Chip
- KRT20, cytokeratin 20
- Muc2, mucin 2
- NHS, N-hydroxysuccinimide
- Olfm4, olfactomedin-4
- P, passage
- PBS, phosphate-buffered saline
- PDMS, polydimethylsiloxane
- PTFE, polytetrafluoroethylene
- Polarized Crypt
- SCFA, short-chain fatty acid
- SEM, scanning electron microscope
- SM, stem medium
- Stem Cell Niche
- TNF-α, tumor necrosis factor-α
- ZO-1, zonula occludens-1
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Raehyun Kim
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Mark I. Reed
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew DiSalvo
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Daniel L. Nguyen
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
29
|
Advancing Intestinal Organoid Technology Toward Regenerative Medicine. Cell Mol Gastroenterol Hepatol 2017; 5:51-60. [PMID: 29204508 PMCID: PMC5704126 DOI: 10.1016/j.jcmgh.2017.10.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
Abstract
With the emergence of technologies to culture intestinal epithelial cells in vitro as various forms of intestinal organoids, there is growing interest in using such cultured intestinal cells as a source for tissue engineering and regenerative medicine. One such approach would be to combine the organoid technology with methodologies to engineer the culture environment, particularly the three-dimensional scaffold materials, to generate intestines that exquisitely recapitulate their original structures and functions. Another approach to use organoids for regenerative medicine would be to urge them to mature into functional intestines by implanting them into hosts. This process includes the tissue-engineered small intestine that uses synthetic scaffolds for tissue regeneration and direct organoid transplantation that takes advantage of submucosal tissues in the native intestines as a scaffold. Further study in these subfields could lead to the development of therapeutic options to use different types of organoids with various cell types in regenerative medicine for intestinal diseases in humans.
Collapse
|
30
|
Wang Y, Gunasekara DB, Attayek PJ, Reed MI, DiSalvo M, Nguyen DL, Dutton JS, Lebhar MS, Bultman SJ, Sims CE, Magness ST, Allbritton NL. In Vitro Generation of Mouse Colon Crypts. ACS Biomater Sci Eng 2017; 3:2502-2513. [PMID: 30854421 DOI: 10.1021/acsbiomaterials.7b00368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Organoid culture has had a significant impact on in vitro studies of the intestinal epithelium; however, the exquisite architecture, luminal accessibility, and lineage compartmentalization found in vivo has not been recapitulated in the organoid systems. We have used a microengineered platform with suitable extracellular matrix contacts and stiffness to generate a self-renewing mouse colonic epithelium that replicates key architectural and physiological functions found in vivo, including a surface lined with polarized crypts. Chemical gradients applied to the basal-luminal axis compartmentalized the stem/progenitor cells and promoted appropriate lineage differentiation along the in vitro crypt axis so that the tissue possessed a crypt stem cell niche as well as a layer of differentiated cells covering the luminal surface. This new approach combining microengineered scaffolds, native chemical gradients, and biophysical cues to control primary epithelium ex vivo can serve as a highly functional and physiologically relevant in vitro tissue model.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Dulan B Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Peter J Attayek
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Mark I Reed
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Matthew DiSalvo
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Daniel L Nguyen
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Johanna S Dutton
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Michael S Lebhar
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Scott J Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Christopher E Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Scott T Magness
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
31
|
Abstract
Recent advances allow access to human cell-based intestinal organoids that recreate human physiology to levels not possible with conventional 2D cell cultures. Despite their huge potential, there are many challenges that remain. This review will cover recent bioengineering approaches to improve organoid maturation, scale up, reproducibility and analysis. The first section covers the advances in engineering the culture environment, followed by the section on tools for micro-manipulation and analysis of organoids. The last section reviews the computational models developed to guide the use of engineered materials and tools, and to interpret observed results as well. The ability to use organoids for discovery research, and the need to both exert exquisite experimental control and obtain quantitative measurements from organoid models means that the field is ripe for collaborative efforts between biologists, engineers, clinicians and industry.
Collapse
Affiliation(s)
- Ge-Ah Kim
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Shuichi Takayama
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
|
33
|
Wang Y, DiSalvo M, Gunasekara DB, Dutton J, Proctor A, Lebhar MS, Williamson IA, Speer J, Howard RL, Smiddy NM, Bultman SJ, Sims CE, Magness ST, Allbritton NL. Self-renewing Monolayer of Primary Colonic or Rectal Epithelial Cells. Cell Mol Gastroenterol Hepatol 2017; 4:165-182.e7. [PMID: 29204504 PMCID: PMC5710741 DOI: 10.1016/j.jcmgh.2017.02.011] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/15/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Three-dimensional organoid culture has fundamentally changed the in vitro study of intestinal biology enabling novel assays; however, its use is limited because of an inaccessible luminal compartment and challenges to data gathering in a three-dimensional hydrogel matrix. Long-lived, self-renewing 2-dimensional (2-D) tissue cultured from primary colon cells has not been accomplished. METHODS The surface matrix and chemical factors that sustain 2-D mouse colonic and human rectal epithelial cell monolayers with cell repertoires comparable to that in vivo were identified. RESULTS The monolayers formed organoids or colonoids when placed in standard Matrigel culture. As with the colonoids, the monolayers exhibited compartmentalization of proliferative and differentiated cells, with proliferative cells located near the peripheral edges of growing monolayers and differentiated cells predominated in the central regions. Screening of 77 dietary compounds and metabolites revealed altered proliferation or differentiation of the murine colonic epithelium. When exposed to a subset of the compound library, murine organoids exhibited similar responses to that of the monolayer but with differences that were likely attributable to the inaccessible organoid lumen. The response of the human primary epithelium to a compound subset was distinct from that of both the murine primary epithelium and human tumor cells. CONCLUSIONS This study demonstrates that a self-renewing 2-D murine and human monolayer derived from primary cells can serve as a physiologically relevant assay system for study of stem cell renewal and differentiation and for compound screening. The platform holds transformative potential for personalized and precision medicine and can be applied to emerging areas of disease modeling and microbiome studies.
Collapse
Key Words
- 2-D, two-dimensional
- 3-D, three-dimensional
- ALP, alkaline phosphatase
- CAG, cytomegalovirus enhancer plus chicken actin promoter
- CI, confidence interval
- Colonic Epithelial Cells
- Compound Screening
- ECM, extracellular matrix
- EDU, 5-ethynyl-2′-deoxyuridine
- EGF, epidermal growth factor
- ENR-W, cell medium with [Wnt-3A] of 30 ng/mL
- ENR-w, cell medium with [Wnt-3A] of 10 ng/mL
- HISC, human intestinal stem cell medium
- IACUC, Institutional Animal Care and Use Committee
- ISC, intestinal stem cell
- Monolayer
- Organoids
- PBS, phosphate-buffered saline
- PDMS, polydimethylsiloxane
- RFP, red fluorescent protein
- SEM, scanning electron microscope
- SSMD, strictly standardized mean difference
- UNC, University of North Carolina
- α-ChgA, anti-chromogranin A
- α-Muc2, anti-mucin2
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew DiSalvo
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Johanna Dutton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Angela Proctor
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Michael S. Lebhar
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Ian A. Williamson
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Riley L. Howard
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina
| | - Nicole M. Smiddy
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina,Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina,Correspondence Address correspondence to: Nancy L. Allbritton, MD, PhD, Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599. fax: (919) 962-2388.Department of ChemistryUniversity of North CarolinaChapel HillNorth Carolina 27599
| |
Collapse
|
34
|
Wang B, Yao M, Lv L, Ling Z, Li L. The Human Microbiota in Health and Disease. ENGINEERING 2017; 3:71-82. [DOI: 10.1016/j.eng.2017.01.008] [Citation(s) in RCA: 460] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
35
|
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
|
36
|
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
|
37
|
Richmond CA, Shah MS, Carlone DL, Breault DT. An enduring role for quiescent stem cells. Dev Dyn 2016; 245:718-26. [PMID: 27153394 DOI: 10.1002/dvdy.24416] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/22/2016] [Accepted: 04/30/2016] [Indexed: 12/12/2022] Open
Abstract
The intestine's ability to recover from catastrophic injury requires quiescent intestinal stem cells (q-ISCs). While rapidly cycling (Lgr5+) crypt base columnar (CBC) ISCs normally maintain the intestine, they are highly sensitive to pathological injuries (irradiation, inflammation) and must be restored by q-ISCs to sustain intestinal homeostasis. Despite clear relevance to human health, virtually nothing is known regarding the factors that regulate q-ISCs. A comprehensive understanding of these mechanisms would likely lead to targeted new therapies with profound therapeutic implications for patients with gastrointestinal conditions. We briefly review the current state of the literature, highlighting homeostatic mechanisms important for q-ISC regulation, listing key questions in the field, and offer strategies to address them. Developmental Dynamics 245:718-726, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Camilla A Richmond
- Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Manasvi S Shah
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts
| | - Diana L Carlone
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
38
|
Kumar A, Starly B. Large scale industrialized cell expansion: producing the critical raw material for biofabrication processes. Biofabrication 2015; 7:044103. [DOI: 10.1088/1758-5090/7/4/044103] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
39
|
Yu J, Cilfone NA, Large EM, Sarkar U, Wishnok JS, Tannenbaum SR, Hughes DJ, Lauffenburger DA, Griffith LG, Stokes CL, Cirit M. Quantitative Systems Pharmacology Approaches Applied to Microphysiological Systems (MPS): Data Interpretation and Multi-MPS Integration. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:585-94. [PMID: 26535159 PMCID: PMC4625863 DOI: 10.1002/psp4.12010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/09/2015] [Indexed: 01/31/2023]
Abstract
Our goal in developing Microphysiological Systems (MPS) technology is to provide an improved approach for more predictive preclinical drug discovery via a highly integrated experimental/computational paradigm. Success will require quantitative characterization of MPSs and mechanistic analysis of experimental findings sufficient to translate resulting insights from in vitro to in vivo. We describe herein a systems pharmacology approach to MPS development and utilization that incorporates more mechanistic detail than traditional pharmacokinetic/pharmacodynamic (PK/PD) models. A series of studies illustrates diverse facets of our approach. First, we demonstrate two case studies: a PK data analysis and an inflammation response--focused on a single MPS, the liver/immune MPS. Building on the single MPS modeling, a theoretical investigation of a four-MPS interactome then provides a quantitative way to consider several pharmacological concepts such as absorption, distribution, metabolism, and excretion in the design of multi-MPS interactome operation and experiments.
Collapse
Affiliation(s)
- J Yu
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - N A Cilfone
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - E M Large
- CN Bio Innovations Welwyn Garden City, UK
| | - U Sarkar
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - J S Wishnok
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - S R Tannenbaum
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA ; Department of Chemistry, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - D J Hughes
- CN Bio Innovations Welwyn Garden City, UK
| | - D A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - L G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA ; Center of Gynepathology, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - C L Stokes
- Stokes Consulting Redwood City, California, USA
| | - M Cirit
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| |
Collapse
|
40
|
In vitro epithelial organoid generation induced by substrate nanotopography. Sci Rep 2015; 5:9293. [PMID: 25787017 PMCID: PMC4365384 DOI: 10.1038/srep09293] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 02/27/2015] [Indexed: 12/21/2022] Open
Abstract
The extracellular matrix (ECM) exhibits tissue-specific topography and composition and plays a crucial role in initiating the biochemical and biomechanical signaling required for organizing cells into distinct tissues during development. How single cells assemble into structures featuring specific shapes in response to external cues is poorly understood. We examined the effect of substrate nanotopography on the morphogenesis of several types of epithelial cells and found that in response to the topography, Calu-3 and MDCK-II cells formed organoids that closely resemble their morphology in vivo. This finding represents the first demonstration that substrate nanotopography, one of the first physical cues detected by cells, can by itself induce epithelial tissue-like organization. Our results provide insights, in terms of a new aspect of ECM topography, into the design of future tissue-engineering systems and the study of mechanosignaling in the epithelium during normal development and tumor progression.
Collapse
|
41
|
Srinivasan B, Kolli AR, Esch MB, Abaci HE, Shuler ML, Hickman JJ. TEER measurement techniques for in vitro barrier model systems. ACTA ACUST UNITED AC 2015; 20:107-26. [PMID: 25586998 DOI: 10.1177/2211068214561025] [Citation(s) in RCA: 1318] [Impact Index Per Article: 131.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transepithelial/transendothelial electrical resistance (TEER) is a widely accepted quantitative technique to measure the integrity of tight junction dynamics in cell culture models of endothelial and epithelial monolayers. TEER values are strong indicators of the integrity of the cellular barriers before they are evaluated for transport of drugs or chemicals. TEER measurements can be performed in real time without cell damage and generally are based on measuring ohmic resistance or measuring impedance across a wide spectrum of frequencies. The measurements for various cell types have been reported with commercially available measurement systems and also with custom-built microfluidic implementations. Some of the barrier models that have been widely characterized using TEER include the blood-brain barrier (BBB), gastrointestinal (GI) tract, and pulmonary models. Variations in these values can arise due to factors such as temperature, medium formulation, and passage number of cells. The aim of this article is to review the different TEER measurement techniques and analyze their strengths and weaknesses, determine the significance of TEER in drug toxicity studies, examine the various in vitro models and microfluidic organs-on-chips implementations using TEER measurements in some widely studied barrier models (BBB, GI tract, and pulmonary), and discuss the various factors that can affect TEER measurements.
Collapse
Affiliation(s)
- Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | - Aditya Reddy Kolli
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | | | | | | | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
42
|
Kurashina Y, Takemura K, Miyata S, Komotori J, Koyama T. Effective cell collection method using collagenase and ultrasonic vibration. BIOMICROFLUIDICS 2014; 8:054118. [PMID: 25584115 PMCID: PMC4290568 DOI: 10.1063/1.4899054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/10/2014] [Indexed: 06/04/2023]
Abstract
This study proposes a novel cell collection method based on collagenase treatment and ultrasonic vibration. The method collects calf chondrocytes from a reusable metal cell culture substrate. To develop our concept, we calculated the natural vibration modes of the cell culture substrate by a finite element method, and conducted eigenvalue and piezoelectric-structural analyses. Selecting the first out-of-plane vibration mode of the substrate, which has a single nodal circle, we designed and fabricated the cell collection device. The excited vibration mode properly realized our intentions. We then evaluated the cell collection ratio and the growth response, and observed the morphology of the collected cells. The collagenase and ultrasonic vibration treatment collected comparable numbers of cells to conventional trypsin and pipetting treatment, but improved the proliferating cell statistics. Morphological observations revealed that the membranes of cells collected by the proposed method remain intact; consequently, the cells are larger and rougher than cells collected by the conventional method. Therefore, we present a promising cell collection method for adhesive cell culturing process.
Collapse
Affiliation(s)
- Y Kurashina
- School of Science for Open and Environmental Systems, Graduate School of Science and Technology, Keio University , Yokohama 223-8522, Japan
| | - K Takemura
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University , Yokohama 223-8522, Japan
| | - S Miyata
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University , Yokohama 223-8522, Japan
| | - J Komotori
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University , Yokohama 223-8522, Japan
| | - T Koyama
- Medicalscience Co. Ltd. , Yokohama 231-0033, Japan
| |
Collapse
|
43
|
Gracz AD, Magness ST. Defining hierarchies of stemness in the intestine: evidence from biomarkers and regulatory pathways. Am J Physiol Gastrointest Liver Physiol 2014; 307:G260-73. [PMID: 24924746 PMCID: PMC4121637 DOI: 10.1152/ajpgi.00066.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
For decades, the rapid proliferation and well-defined cellular lineages of the small intestinal epithelium have driven an interest in the biology of the intestinal stem cells (ISCs) and progenitors that produce the functional cells of the epithelium. Recent and significant advances in ISC biomarker discovery have established the small intestinal epithelium as a powerful model system for studying general paradigms in somatic stem cell biology and facilitated elegant genetic and functional studies of stemness in the intestine. However, this newfound wealth of ISC biomarkers raises important questions of marker specificity. Furthermore, the ISC field must now begin to reconcile biomarker status with functional stemness, a challenge that is made more complex by emerging evidence that cellular hierarchies in the intestinal epithelium are more plastic than previously imagined, with some progenitor populations capable of dedifferentiating and functioning as ISCs following damage. In this review, we discuss the state of the ISC field in terms of biomarkers, tissue dynamics, and cellular hierarchies, and how these processes might be informed by earlier studies into signaling networks in the small intestine.
Collapse
Affiliation(s)
- A. D. Gracz
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; ,2Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - S. T. Magness
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; ,2Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and ,3Joint Department of Biomedical Engineering, University of North Carolina/North Carolina State University, Chapel Hill, North Carolina
| |
Collapse
|