151
|
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
|
152
|
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
|
153
|
Sharma A, Raman V, Lee J, Forbes NS. Mucus blocks probiotics but increases penetration of motile pathogens and induces TNF-α and IL-8 secretion. Biotechnol Bioeng 2020; 117:2540-2555. [PMID: 32396232 PMCID: PMC7806204 DOI: 10.1002/bit.27383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/29/2022]
Abstract
The mucosal barrier in combination with innate immune system are the first line of defense against luminal bacteria at the intestinal mucosa. Dysfunction of the mucus layer and bacterial infiltration are linked to tissue inflammation and disease. To study host-bacterial interactions at the mucosal interface, we created an experimental model that contains luminal space, a mucus layer, an epithelial layer, and suspended immune cells. Reconstituted porcine small intestinal mucus formed an 880 ± 230 µm thick gel layer and had a porous structure. In the presence of mucus, sevenfold less probiotic and nonmotile VSL#3 bacteria transmigrated across the epithelial barrier compared to no mucus. The higher bacterial transmigration caused immune cell differentiation and increased the concentration of interleukin-8 (IL-8) and tumor necrosis factor-alpha (TNF-α; p < .01). Surprisingly, the mucus layer increased transmigration of pathogenic Salmonella and increased secretion of TNF-α and IL-8 (p < .05). Nonmotile, flagella knockout Salmonella had lower transmigration and caused lower IL-8 and TNF-α secretion (p < .05). These results demonstrate that motility enables pathogenic bacteria to cross the mucus and epithelial layers, which could lead to infection. Using an in vitro coculture platform to understand the interactions of bacteria with the intestinal mucosa has the potential to improve the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|
154
|
Steinway SN, Saleh J, Koo BK, Delacour D, Kim DH. Human Microphysiological Models of Intestinal Tissue and Gut Microbiome. Front Bioeng Biotechnol 2020; 8:725. [PMID: 32850690 PMCID: PMC7411353 DOI: 10.3389/fbioe.2020.00725] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is a complex system responsible for nutrient absorption, digestion, secretion, and elimination of waste products that also hosts immune surveillance, the intestinal microbiome, and interfaces with the nervous system. Traditional in vitro systems cannot harness the architectural and functional complexity of the GI tract. Recent advances in organoid engineering, microfluidic organs-on-a-chip technology, and microfabrication allows us to create better in vitro models of human organs/tissues. These micro-physiological systems could integrate the numerous cell types involved in GI development and physiology, including intestinal epithelium, endothelium (vascular), nerve cells, immune cells, and their interplay/cooperativity with the microbiome. In this review, we report recent progress in developing micro-physiological models of the GI systems. We also discuss how these models could be used to study normal intestinal physiology such as nutrient absorption, digestion, and secretion as well as GI infection, inflammation, cancer, and metabolism.
Collapse
Affiliation(s)
- Steven N. Steinway
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jad Saleh
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology, Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Delphine Delacour
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Deok-Ho Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
155
|
Abstract
Organoids form through self-organization processes in which initially homogeneous populations of stem cells spontaneously break symmetry and undergo in-vivo-like pattern formation and morphogenesis, though the processes controlling this are poorly characterized. While these in vitro self-organized tissues far exceed the microscopic and functional complexity obtained by current tissue engineering technologies, they are non-physiological in shape and size and have limited function and lifespan. Here, we discuss how engineering efforts for guiding stem-cell-based development at multiple stages can form the basis for the assembly of highly complex and rationally designed self-organizing multicellular systems with increased robustness and physiological relevance.
Collapse
|
156
|
Shin YC, Shin W, Koh D, Wu A, Ambrosini YM, Min S, Eckhardt SG, Fleming RYD, Kim S, Park S, Koh H, Yoo TK, Kim HJ. Three-Dimensional Regeneration of Patient-Derived Intestinal Organoid Epithelium in a Physiodynamic Mucosal Interface-on-a-Chip. MICROMACHINES 2020; 11:E663. [PMID: 32645991 PMCID: PMC7408321 DOI: 10.3390/mi11070663] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/29/2022]
Abstract
The regeneration of the mucosal interface of the human intestine is critical in the host-gut microbiome crosstalk associated with gastrointestinal diseases. The biopsy-derived intestinal organoids provide genetic information of patients with physiological cytodifferentiation. However, the enclosed lumen and static culture condition substantially limit the utility of patient-derived organoids for microbiome-associated disease modeling. Here, we report a patient-specific three-dimensional (3D) physiodynamic mucosal interface-on-a-chip (PMI Chip) that provides a microphysiological intestinal milieu under defined biomechanics. The real-time imaging and computational simulation of the PMI Chip verified the recapitulation of non-linear luminal and microvascular flow that simulates the hydrodynamics in a living human gut. The multiaxial deformations in a convoluted microchannel not only induced dynamic cell strains but also enhanced particle mixing in the lumen microchannel. Under this physiodynamic condition, an organoid-derived epithelium obtained from the patients diagnosed with Crohn's disease, ulcerative colitis, or colorectal cancer independently formed 3D epithelial layers with disease-specific differentiations. Moreover, co-culture with the human fecal microbiome in an anoxic-oxic interface resulted in the formation of stochastic microcolonies without a loss of epithelial barrier function. We envision that the patient-specific PMI Chip that conveys genetic, epigenetic, and environmental factors of individual patients will potentially demonstrate the pathophysiological dynamics and complex host-microbiome crosstalk to target a patient-specific disease modeling.
Collapse
Affiliation(s)
- Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Domin Koh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Alexander Wu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Yoko M. Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - S. Gail Eckhardt
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
| | - R. Y. Declan Fleming
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Seung Kim
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Sowon Park
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Hong Koh
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Tae Kyung Yoo
- Department of Computer Art, College of Art and Technology, Chung-Ang University, Seoul 06974, Korea;
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
- Department of Medical Engineering, College of Medicine, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
157
|
Huang WK, Xie C, Young RL, Zhao JB, Ebendorff-Heidepriem H, Jones KL, Rayner CK, Wu TZ. Development of innovative tools for investigation of nutrient-gut interaction. World J Gastroenterol 2020; 26:3562-3576. [PMID: 32742126 PMCID: PMC7366065 DOI: 10.3748/wjg.v26.i25.3562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract is the key interface between the ingesta and the human body. There is wide recognition that the gastrointestinal response to nutrients or bioactive compounds, particularly the secretion of numerous hormones, is critical to the regulation of appetite, body weight and blood glucose. This concept has led to an increasing focus on “gut-based” strategies for the management of metabolic disorders, including type 2 diabetes and obesity. Understanding the underlying mechanisms and downstream effects of nutrient-gut interactions is fundamental to effective translation of this knowledge to clinical practice. To this end, an array of research tools and platforms have been developed to better understand the mechanisms of gut hormone secretion from enteroendocrine cells. This review discusses the evolution of in vitro and in vivo models and the integration of innovative techniques that will ultimately enable the development of novel therapies for metabolic diseases.
Collapse
Affiliation(s)
- Wei-Kun Huang
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Institute for Photonics and Advanced Sensing, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
- The ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Cong Xie
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
| | - Richard L Young
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Diabetes, Nutrition and Gut Health, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, SA 5005, Australia
| | - Jiang-Bo Zhao
- Institute for Photonics and Advanced Sensing, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
- The ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Heike Ebendorff-Heidepriem
- Institute for Photonics and Advanced Sensing, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
- The ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Karen L Jones
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
| | - Christopher K Rayner
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Tong-Zhi Wu
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
158
|
D'Costa K, Kosic M, Lam A, Moradipour A, Zhao Y, Radisic M. Biomaterials and Culture Systems for Development of Organoid and Organ-on-a-Chip Models. Ann Biomed Eng 2020; 48:2002-2027. [PMID: 32285341 PMCID: PMC7334104 DOI: 10.1007/s10439-020-02498-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
The development of novel 3D tissue culture systems has enabled the in vitro study of in vivo processes, thereby overcoming many of the limitations of previous 2D tissue culture systems. Advances in biomaterials, including the discovery of novel synthetic polymers has allowed for the generation of physiologically relevant in vitro 3D culture models. A large number of 3D culture systems, aided by novel organ-on-a-chip and bioreactor technologies have been developed to improve reproducibility and scalability of in vitro organ models. The discovery of induced pluripotent stem cells (iPSCs) and the increasing number of protocols to generate iPSC-derived cell types has allowed for the generation of novel 3D models with minimal ethical limitations. The production of iPSC-derived 3D cultures has revolutionized the field of developmental biology and in particular, the study of fetal brain development. Furthermore, physiologically relevant 3D cultures generated from PSCs or adult stem cells (ASCs) have greatly advanced in vitro disease modelling and drug discovery. This review focuses on advances in 3D culture systems over the past years to model fetal development, disease pathology and support drug discovery in vitro, with a specific focus on the enabling role of biomaterials.
Collapse
Affiliation(s)
- Katya D'Costa
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Milena Kosic
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Angus Lam
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Azeen Moradipour
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
159
|
Yavitt FM, Brown TE, Hushka EA, Brown ME, Gjorevski N, Dempsey PJ, Lutolf MP, Anseth KS. The Effect of Thiol Structure on Allyl Sulfide Photodegradable Hydrogels and their Application as a Degradable Scaffold for Organoid Passaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1905366. [PMID: 32548863 PMCID: PMC7669673 DOI: 10.1002/adma.201905366] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 04/19/2020] [Indexed: 05/17/2023]
Abstract
Intestinal organoids are useful in vitro models for basic and translational studies aimed at understanding and treating disease. However, their routine culture relies on animal-derived matrices that limit translation to clinical applications. In fact, there are few fully defined, synthetic hydrogel systems that allow for the expansion of intestinal organoids. Here, an allyl sulfide photodegradable hydrogel is presented, achieving rapid degradation through radical addition-fragmentation chain transfer (AFCT) reactions, to support routine passaging of intestinal organoids. Shear rheology to first characterize the effect of thiol and allyl sulfide crosslink structures on degradation kinetics is used. Irradiation with 365 nm light (5 mW cm-2 ) in the presence of a soluble thiol (glutathione at 15 × 10-3 m), and a photoinitiator (lithium phenyl-2,4,6-trimethylbenzoylphosphinate at 1 × 10-3 m), leads to complete hydrogel degradation in less than 15 s. Allyl sulfide hydrogels are used to support the formation of epithelial colonies from single intestinal stem cells, and rapid photodegradation is used to achieve repetitive passaging of stem cell colonies without loss in morphology or organoid formation potential. This platform could support long-term culture of intestinal organoids, potentially replacing the need for animal-derived matrices, while also allowing systematic variations to the hydrogel properties tailored for the organoid of interest.
Collapse
Affiliation(s)
- F. Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Tobin E. Brown
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Current address: Material Measurement Laboratory, National Institute of Standards and Technology, Boulder, CO 80305, USA
| | - Ella A. Hushka
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Monica E. Brown
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Denver, CO 80204, USA
| | - Nikolche Gjorevski
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peter J. Dempsey
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Denver, CO 80204, USA
| | - Matthias P. Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), EPFL
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
160
|
Caruso G, Musso N, Grasso M, Costantino A, Lazzarino G, Tascedda F, Gulisano M, Lunte SM, Caraci F. Microfluidics as a Novel Tool for Biological and Toxicological Assays in Drug Discovery Processes: Focus on Microchip Electrophoresis. MICROMACHINES 2020; 11:E593. [PMID: 32549277 PMCID: PMC7344675 DOI: 10.3390/mi11060593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
The last decades of biological, toxicological, and pharmacological research have deeply changed the way researchers select the most appropriate 'pre-clinical model'. The absence of relevant animal models for many human diseases, as well as the inaccurate prognosis coming from 'conventional' pre-clinical models, are among the major reasons of the failures observed in clinical trials. This evidence has pushed several research groups to move more often from a classic cellular or animal modeling approach to an alternative and broader vision that includes the involvement of microfluidic-based technologies. The use of microfluidic devices offers several benefits including fast analysis times, high sensitivity and reproducibility, the ability to quantitate multiple chemical species, and the simulation of cellular response mimicking the closest human in vivo milieu. Therefore, they represent a useful way to study drug-organ interactions and related safety and toxicity, and to model organ development and various pathologies 'in a dish'. The present review will address the applicability of microfluidic-based technologies in different systems (2D and 3D). We will focus our attention on applications of microchip electrophoresis (ME) to biological and toxicological studies as well as in drug discovery and development processes. These include high-throughput single-cell gene expression profiling, simultaneous determination of antioxidants and reactive oxygen and nitrogen species, DNA analysis, and sensitive determination of neurotransmitters in biological fluids. We will discuss new data obtained by ME coupled to laser-induced fluorescence (ME-LIF) and electrochemical detection (ME-EC) regarding the production and degradation of nitric oxide, a fundamental signaling molecule regulating virtually every critical cellular function. Finally, the integration of microfluidics with recent innovative technologies-such as organoids, organ-on-chip, and 3D printing-for the design of new in vitro experimental devices will be presented with a specific attention to drug development applications. This 'composite' review highlights the potential impact of 2D and 3D microfluidic systems as a fast, inexpensive, and highly sensitive tool for high-throughput drug screening and preclinical toxicological studies.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy; (N.M.); (G.L.)
| | - Margherita Grasso
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| | - Angelita Costantino
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy; (N.M.); (G.L.)
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Massimo Gulisano
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
- Molecular Preclinical and Translational Imaging Research Centre-IMPRonTE, University of Catania, 95125 Catania, Italy
- Interuniversity Consortium for Biotechnology, Area di Ricerca, Padriciano, 34149 Trieste, Italy
| | - Susan M. Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA;
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| |
Collapse
|
161
|
Zhang M, Liu Y, Chen YG. Generation of 3D human gastrointestinal organoids: principle and applications. ACTA ACUST UNITED AC 2020; 9:6. [PMID: 32588198 PMCID: PMC7306834 DOI: 10.1186/s13619-020-00040-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
The stomach and intestine are important organs for food digestion, nutrient absorption, immune protection and hormone production. Gastrointestinal diseases such as cancer and ulcer are big threats to human health. Appropriate disease models are in sore need for mechanistic understanding and drug discovery. Organoids are three-dimensional in vitro cultured structures derived from tissues and pluripotent stem cells with multiple types of cells and mimicking in vivo tissues in major aspects. They have a great potential in regenerative medicine and personalized medicine. Here, we review the major signaling pathways regulating gastrointestinal epithelial homeostasis, summarize different methods to generate human gastrointestinal organoids and highlight their applications in biological research and medical practice.
Collapse
Affiliation(s)
- Mengxian Zhang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
162
|
Mechanical Adaptations of Epithelial Cells on Various Protruded Convex Geometries. Cells 2020; 9:cells9061434. [PMID: 32527037 PMCID: PMC7349491 DOI: 10.3390/cells9061434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
The shape of epithelial tissue supports physiological functions of organs such as intestinal villi and corneal epithelium. Despite the mounting evidence showing the importance of geometry in tissue microenvironments, the current understanding on how it affects biophysical behaviors of cells is still elusive. Here, we cultured cells on various protruded convex structure such as triangle, square, and circle shape fabricated using two-photon laser lithography and quantitatively analyzed individual cells. Morphological data indicates that epithelial cells can sense the sharpness of the corner by showing the characteristic cell alignments, which was caused by actin contractility. Cell area was mainly influenced by surface convexity, and Rho-activation increased cell area on circle shape. Moreover, we found that intermediate filaments, vimentin, and cytokeratin 8/18, play important roles in growth and adaptation of epithelial cells by enhancing expression level on convex structure depending on the shape. In addition, microtubule building blocks, α-tubulin, was also responded on geometric structure, which indicates that intermediate filaments and microtubule can cooperatively secure mechanical stability of epithelial cells on convex surface. Altogether, the current study will expand our understanding of mechanical adaptations of cells on out-of-plane geometry.
Collapse
|
163
|
High-throughput automated organoid culture via stem-cell aggregation in microcavity arrays. Nat Biomed Eng 2020; 4:863-874. [PMID: 32514094 DOI: 10.1038/s41551-020-0565-2] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/29/2020] [Indexed: 11/09/2022]
Abstract
Stem-cell-derived epithelial organoids are routinely used for the biological and biomedical modelling of tissues. However, the complexity, lack of standardization and quality control of stem cell culture in solid extracellular matrices hampers the routine use of the organoids at the industrial scale. Here, we report the fabrication of microengineered cell culture devices and scalable and automated methods for suspension culture and real-time analysis of thousands of individual gastrointestinal organoids trapped in microcavity arrays within a polymer-hydrogel substrate. The absence of a solid matrix substantially reduces organoid heterogeneity, which we show for mouse and human gastrointestinal organoids. We use the devices to screen for anticancer drug candidates with patient-derived colorectal cancer organoids, and apply high-content image-based phenotypic analyses to reveal insights into mechanisms of drug action. The scalable organoid-culture technology should facilitate the use of organoids in drug development and diagnostics.
Collapse
|
164
|
Shin W, Hackley LA, Kim HJ. "Good Fences Make Good Neighbors": How does the Human Gut Microchip Unravel Mechanism of Intestinal Inflammation? Gut Microbes 2020; 11:581-586. [PMID: 31198078 PMCID: PMC7524309 DOI: 10.1080/19490976.2019.1626684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A microengineered human gut-on-a-chip has demonstrated intestinal physiology, three-dimensional (3D) epithelial morphogenesis, and longitudinal host-microbiome interactions in vitro. The modular accessibility and modularity of the microphysiological gut-on-a-chip can lead to the identification of the seminal trigger in intestinal inflammation. By coupling microbial and immune cells in a spatiotemporal manner, we discovered that the maintenance of healthy epithelial barrier function is necessary and sufficient to demonstrate the homeostatic tolerance of the gut. Here, we highlight the breakthrough of our new disease model and discuss the future impact of investigating the etiology and therapeutic targets in the multifactorial inflammatory bowel disease.
Collapse
Affiliation(s)
- Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Landon A. Hackley
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA,CONTACT Hyun Jung Kim Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton St., BME 4.202C, Austin, TX78712, USA
| |
Collapse
|
165
|
Altay G, Tosi S, García-Díaz M, Martínez E. Imaging the Cell Morphological Response to 3D Topography and Curvature in Engineered Intestinal Tissues. Front Bioeng Biotechnol 2020; 8:294. [PMID: 32318564 PMCID: PMC7154059 DOI: 10.3389/fbioe.2020.00294] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/19/2020] [Indexed: 12/11/2022] Open
Abstract
While conventional cell culture methodologies have relied on flat, two-dimensional cell monolayers, three-dimensional engineered tissues are becoming increasingly popular. Often, engineered tissues can mimic the complex architecture of native tissues, leading to advancements in reproducing physiological functional properties. In particular, engineered intestinal tissues often use hydrogels to mimic villi structures. These finger-like protrusions of a few hundred microns in height have a well-defined topography and curvature. Here, we examined the cell morphological response to these villus-like microstructures at single-cell resolution using a novel embedding method that allows for the histological processing of these delicate hydrogel structures. We demonstrated that by using photopolymerisable poly(ethylene) glycol as an embedding medium, the villus-like microstructures were successfully preserved after sectioning with vibratome or cryotome. Moreover, high-resolution imaging of these sections revealed that cell morphology, nuclei orientation, and the expression of epithelial polarization markers were spatially encoded along the vertical axis of the villus-like microstructures and that this cell morphological response was dramatically affected by the substrate curvature. These findings, which are in good agreement with the data reported for in vivo experiments on the native tissue, are likely to be the origin of more physiologically relevant barrier properties of engineered intestinal tissues when compared with standard monolayer cultures. By showcasing this example, we anticipate that the novel histological embedding procedure will have a positive impact on the study of epithelial cell behavior on three-dimensional substrates in both physiological and pathological situations.
Collapse
Affiliation(s)
- Gizem Altay
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sébastien Tosi
- Advanced Digital Microscopy Core Facility, Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - María García-Díaz
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Elena Martínez
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
166
|
Peters MF, Choy AL, Pin C, Leishman DJ, Moisan A, Ewart L, Guzzie-Peck PJ, Sura R, Keller DA, Scott CW, Kolaja KL. Developing in vitro assays to transform gastrointestinal safety assessment: potential for microphysiological systems. LAB ON A CHIP 2020; 20:1177-1190. [PMID: 32129356 DOI: 10.1039/c9lc01107b] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Drug-induced gastrointestinal toxicities (DI-GITs) are among the most common adverse events in clinical trials. High prevalence of DI-GIT has persisted among new drugs due in part to the lack of robust experimental tools to allow early detection or to guide optimization of safer molecules. Developing in vitro assays for the leading GI toxicities (nausea, vomiting, diarrhoea, constipation, and abdominal pain) will likely involve recapitulating complex physiological properties that require contributions from diverse cell/tissue types including epithelial, immune, microbiome, nerve, and muscle. While this stipulation may be beyond traditional 2D monocultures of intestinal cell lines, emerging 3D GI microtissues capture interactions between diverse cell and tissue types. These interactions give rise to microphysiologies fundamental to gut biology. For GI microtissues, organoid technology was the breakthrough that introduced intestinal stem cells with the capability of differentiating into each of the epithelial cell types and that self-organize into a multi-cellular tissue proxy with villus- and crypt-like domains. Recently, GI microtissues generated using miniaturized devices with microfluidic flow and cyclic peristaltic strain were shown to induce Caco2 cells to spontaneously differentiate into each of the principle intestinal epithelial cell types. Second generation models comprised of epithelial organoids or microtissues co-cultured with non-epithelial cell types can successfully reproduce cross-'tissue' functional interactions broadening the potential of these models to accurately study drug-induced toxicities. A new paradigm in which in vitro assays become an early part of GI safety assessment could be realized if microphysiological systems (MPS) are developed in alignment with drug-discovery needs. Herein, approaches for assessing GI toxicity of pharmaceuticals are reviewed and gaps are compared with capabilities of emerging GI microtissues (e.g., organoids, organ-on-a-chip, transwell systems) in order to provide perspective on the assay features needed for MPS models to be adopted for DI-GIT assessment.
Collapse
Affiliation(s)
- Matthew F Peters
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Bruce J, Kaiko GE, Keely S. Isolation and In Vitro Culture of Human Gut Progenitor Cells. Methods Mol Biol 2020; 2029:49-62. [PMID: 31273733 DOI: 10.1007/978-1-4939-9631-5_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The gastrointestinal epithelium is a highly regenerative organ, where each cell is replaced in a cycle of 4-6 days, depending on the mammalian species. This highly proliferative state is driven by gastrointestinal stem and progenitor cells, located at the base of crypts. These cells give rise to at least six types of differentiated epithelial cells, each with a distinct function in maintaining homeostasis between the intestinal interface and the luminal environment. The isolation and culture of these cells from mammalian gastrointestinal tissue is a novel technique, which allows for the generation and maintenance of an in vitro culture system for adult epithelial cells. There are two predominant methods for isolation and propagation of gastrointestinal epithelial cells, the first is the organoid system developed in 2009, and the second is a later version known as the L-WRN spheroid system. In this chapter, we describe the method to isolate and culture human gastrointestinal stem and progenitor cells and culture them as three-dimensional spheroids using L-WRN cell conditioned media.
Collapse
Affiliation(s)
- Jessica Bruce
- Viruses, Infections/Immunity, Vaccines and Asthma (VIVA) Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Gerard E Kaiko
- Viruses, Infections/Immunity, Vaccines and Asthma (VIVA) Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Simon Keely
- Viruses, Infections/Immunity, Vaccines and Asthma (VIVA) Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia. .,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
168
|
Pradhan S, Banda OA, Farino CJ, Sperduto JL, Keller KA, Taitano R, Slater JH. Biofabrication Strategies and Engineered In Vitro Systems for Vascular Mechanobiology. Adv Healthc Mater 2020; 9:e1901255. [PMID: 32100473 PMCID: PMC8579513 DOI: 10.1002/adhm.201901255] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The vascular system is integral for maintaining organ-specific functions and homeostasis. Dysregulation in vascular architecture and function can lead to various chronic or acute disorders. Investigation of the role of the vascular system in health and disease has been accelerated through the development of tissue-engineered constructs and microphysiological on-chip platforms. These in vitro systems permit studies of biochemical regulation of vascular networks and parenchymal tissue and provide mechanistic insights into the biophysical and hemodynamic forces acting in organ-specific niches. Detailed understanding of these forces and the mechanotransductory pathways involved is necessary to develop preventative and therapeutic strategies targeting the vascular system. This review describes vascular structure and function, the role of hemodynamic forces in maintaining vascular homeostasis, and measurement approaches for cell and tissue level mechanical properties influencing vascular phenomena. State-of-the-art techniques for fabricating in vitro microvascular systems, with varying degrees of biological and engineering complexity, are summarized. Finally, the role of vascular mechanobiology in organ-specific niches and pathophysiological states, and efforts to recapitulate these events using in vitro microphysiological systems, are explored. It is hoped that this review will help readers appreciate the important, but understudied, role of vascular-parenchymal mechanotransduction in health and disease toward developing mechanotherapeutics for treatment strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Omar A. Banda
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Keely A. Keller
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Ryan Taitano
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
169
|
Jing B, Wang ZA, Zhang C, Deng Q, Wei J, Luo Y, Zhang X, Li J, Du Y. Establishment and Application of Peristaltic Human Gut-Vessel Microsystem for Studying Host-Microbial Interaction. Front Bioeng Biotechnol 2020; 8:272. [PMID: 32296697 PMCID: PMC7137556 DOI: 10.3389/fbioe.2020.00272] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/16/2020] [Indexed: 12/18/2022] Open
Abstract
Intestinal floras influence a lot of biological functions of the organism. Although animal model are strong tools for researches on the relationship between host and microbe, a physiologically relevant in vitro human gut model was still required. Here, a novel human gut-vessel microfluidic system was established to study the host–microbial interaction. Peristaltic motion of the cells on the chip was driven by a pneumatic pump. When intestinal epithelial cells (Caco2) were co-cultured with vascular endothelial cells (HUVECs) on the peristaltic microfluidic chip, Caco2 showed normal barrier and absorption functions after 5 days cultivation, which generally took 21 days in static Transwell models. Intestinal microvilli and glycocalyx layer were seen after 4 days cultivation, and Lactobacillus casei was successfully co-cultured for a week in the intestinal cavity. A model for intestinal damage and inflammatory responses caused by E. coli was set up on this chip, which were successfully suppressed by Lactobacillus casei or antibiotic. In summary, this human gut-vessel microfluidic system showed a good potential for investigating the host–microbial interaction and the effect and mechanism of microbiome on intestinal diseases in vitro.
Collapse
Affiliation(s)
- Bolin Jing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China.,Department of Chemistry, University of Chinese Academy of Sciences, Beijing, China
| | - Zhuo A Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Chen Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Quanfeng Deng
- Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Jinhua Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Yong Luo
- Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Xiuli Zhang
- College of Pharmaceutical Sciences, Soochow University, Soochow, China
| | - Jianjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Yuguang Du
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
170
|
Huang J, Jiang Y, Ren Y, Liu Y, Wu X, Li Z, Ren J. Biomaterials and biosensors in intestinal organoid culture, a progress review. J Biomed Mater Res A 2020; 108:1501-1508. [PMID: 32170907 DOI: 10.1002/jbm.a.36921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
As an emerging technology, intestinal organoids are promising new tools for basic and translational research in gastroenterology. Currently, culture of intestinal organoids relies mostly on a type of tumor-derived scaffolds, namely Matrigel, which may pose tumorigenic risks to organoid implantation. Apart from the traditional detection methods, such as tissue slicing and fluorescence staining, the monitoring of intestinal organoids requires real-time biosensors that can adapt to their three-dimensional dynamic growth patterns. In this review, we summarized the recent advances in developing definite hydrogel scaffolds for intestinal organoid culture and identified key parameters for scaffold design. In addition, classified by different substrate compositions like pH, electrolytes, and functional proteins, we concluded the existing live-imaging biosensors and elucidated their underlying mechanisms. We hope this review enhances the understanding of intestinal organoid culture and provides more practical approaches to investigate them.
Collapse
Affiliation(s)
- Jinjian Huang
- School of Medicine, Southeast University, Nanjing, China.,Laboratory for Trauma and Surgical Infections, Research Institute of General Surgery, Jinling Hospital, Nanjing, China
| | - Yungang Jiang
- School of Medicine, Southeast University, Nanjing, China.,Laboratory for Trauma and Surgical Infections, Research Institute of General Surgery, Jinling Hospital, Nanjing, China
| | - Yanhan Ren
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Ye Liu
- School of Medicine, Southeast University, Nanjing, China.,Laboratory for Trauma and Surgical Infections, Research Institute of General Surgery, Jinling Hospital, Nanjing, China
| | - Xiuwen Wu
- Laboratory for Trauma and Surgical Infections, Research Institute of General Surgery, Jinling Hospital, Nanjing, China
| | - Zongan Li
- Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing, NARI School of Electrical and Automation Engineering, Nanjing Normal University, Nanjing, China
| | - Jianan Ren
- School of Medicine, Southeast University, Nanjing, China.,Laboratory for Trauma and Surgical Infections, Research Institute of General Surgery, Jinling Hospital, Nanjing, China
| |
Collapse
|
171
|
Peters MF, Landry T, Pin C, Maratea K, Dick C, Wagoner MP, Choy AL, Barthlow H, Snow D, Stevens Z, Armento A, Scott CW, Ayehunie S. Human 3D Gastrointestinal Microtissue Barrier Function As a Predictor of Drug-Induced Diarrhea. Toxicol Sci 2020; 168:3-17. [PMID: 30364994 PMCID: PMC6390652 DOI: 10.1093/toxsci/kfy268] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Drug-induced gastrointestinal toxicities (GITs) rank among the most common clinical side effects. Preclinical efforts to reduce incidence are limited by inadequate predictivity of in vitro assays. Recent breakthroughs in in vitro culture methods support intestinal stem cell maintenance and continual differentiation into the epithelial cell types resident in the intestine. These diverse cells self-assemble into microtissues with in vivo-like architecture. Here, we evaluate human GI microtissues grown in transwell plates that allow apical and/or basolateral drug treatment and 96-well throughput. Evaluation of assay utility focused on predictivity for diarrhea because this adverse effect correlates with intestinal barrier dysfunction which can be measured in GI microtissues using transepithelial electrical resistance (TEER). A validation set of widely prescribed drugs was assembled and tested for effects on TEER. When the resulting TEER inhibition potencies were adjusted for clinical exposure, a threshold was identified that distinguished drugs that induced clinical diarrhea from those that lack this liability. Microtissue TEER assay predictivity was further challenged with a smaller set of drugs whose clinical development was limited by diarrhea that was unexpected based on 1-month animal studies. Microtissue TEER accurately predicted diarrhea for each of these drugs. The label-free nature of TEER enabled repeated quantitation with sufficient precision to develop a mathematical model describing the temporal dynamics of barrier damage and recovery. This human 3D GI microtissue is the first in vitro assay with validated predictivity for diarrhea-inducing drugs. It should provide a platform for lead optimization and offers potential for dose schedule exploration.
Collapse
Affiliation(s)
- Matthew F Peters
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Tim Landry
- MatTek Corporation, Ashland, Massachusetts 01721
| | - Carmen Pin
- Mechanistic Safety and ADME Sciences, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Kim Maratea
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Cortni Dick
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Matthew P Wagoner
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Allison L Choy
- Science and Enabling Units IT, AstraZeneca, Waltham, MA 02451
| | - Herb Barthlow
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | - Deb Snow
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | | | - Alex Armento
- MatTek Corporation, Ashland, Massachusetts 01721
| | - Clay W Scott
- Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, MA 02451
| | | |
Collapse
|
172
|
Dosh RH, Jordan-Mahy N, Sammon C, Le Maitre CL. Use of l-pNIPAM hydrogel as a 3D-scaffold for intestinal crypts and stem cell tissue engineering. Biomater Sci 2020; 7:4310-4324. [PMID: 31410428 DOI: 10.1039/c9bm00541b] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Intestinal stem cells hold great potential in tissue regeneration of the intestine, however, there are key limitations in their culture in vitro. We previously reported a novel synthetic non-biodegradable hydrogel as a 3D culture model for intestinal epithelium using Caco2 and HT29-MTX cells. Here, we investigated the potential of this system as a 3D scaffold for crypts and single intestinal stem cells to support long-term culture and differentiation. Intestinal crypts were extracted from murine small intestines and Lgr5+ stem cells isolated by magnetic activated cell sorting. Crypts and stem cells were suspended within Matrigel or l-pNIPAM for 14 days or suspended within Matrigel for 7 days then released, dissociated, and suspended within, or on l-pNIPAM hydrogel for 28 days. Cellular behaviour and phenotype were determined by histology and immunohistochemistry for stem cell and differentiation markers: Lgr5, E-cadherin MUC2 chromograninA and lysozymes. Isolated crypts and Lgr5+ intestinal stem cells formed enteroids with a central lumen surrounded by multiple crypt-like buds when cultured in Matrigel. In contrast, when crypts and stem cells were directly suspended within, or layered on l-pNIPAM hydrogel under dynamic culture conditions they formed spherical balls of cells, with no central lumen. When enteroids were initially formed in Matrigel from crypts or single Lgr5+ intestinal stem cells and dissociated into small fragments or single cells and transferred to l-pNIPAM hydrogel they formed new larger enteroids with numerous crypt-like buds. These crypt-like buds showed the presence of mucin-producing cells, which resembled goblet cells, scattered throughout their structures. Immunohistochemistry staining also showed the expression of Lgr5 and differentiation markers of all the main intestinal cell types including: enterocytes, goblet cells, enteroendocrine and Paneth cells. This demonstrated that l-pNIPAM hydrogel supported long-term culture of crypts and Lgr5+ stem cells and promoted intestinal cell differentiation.
Collapse
Affiliation(s)
- Rasha H Dosh
- Biomolecular Sciences Research Centre, Sheffield Hallam University, S1 1WB, UK.
| | | | | | | |
Collapse
|
173
|
Ao Z, Cai H, Havert DJ, Wu Z, Gong Z, Beggs JM, Mackie K, Guo F. One-Stop Microfluidic Assembly of Human Brain Organoids To Model Prenatal Cannabis Exposure. Anal Chem 2020; 92:4630-4638. [PMID: 32070103 DOI: 10.1021/acs.analchem.0c00205] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Prenatal cannabis exposure (PCE) influences human brain development, but it is challenging to model PCE using animals and current cell culture techniques. Here, we developed a one-stop microfluidic platform to assemble and culture human cerebral organoids from human embryonic stem cells (hESC) to investigate the effect of PCE on early human brain development. By incorporating perfusable culture chambers, air-liquid interface, and one-stop protocol, this microfluidic platform can simplify the fabrication procedure and produce a large number of organoids (169 organoids per 3.5 cm × 3.5 cm device area) without fusion, as compared with conventional fabrication methods. These one-stop microfluidic assembled cerebral organoids not only recapitulate early human brain structure, biology, and electrophysiology but also have minimal size variation and hypoxia. Under on-chip exposure to the psychoactive cannabinoid, Δ-9-tetrahydrocannabinol (THC), cerebral organoids exhibited reduced neuronal maturation, downregulation of cannabinoid receptor type 1 (CB1) receptors, and impaired neurite outgrowth. Moreover, transient on-chip THC treatment also decreased spontaneous firing in these organoids. This one-stop microfluidic technique enables a simple, scalable, and repeatable organoid culture method that can be used not only for human brain organoids but also for many other human organoids including liver, kidney, retina, and tumor organoids. This technology could be widely used in modeling brain and other organ development, developmental disorders, developmental pharmacology and toxicology, and drug screening.
Collapse
Affiliation(s)
- Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Daniel J Havert
- Department of Physics, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhiyi Gong
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - John M Beggs
- Department of Physics, Indiana University, Bloomington, Indiana 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
174
|
Shafiee A. Design and Fabrication of Three-Dimensional Printed Scaffolds for Cancer Precision Medicine. Tissue Eng Part A 2020; 26:305-317. [PMID: 31992154 DOI: 10.1089/ten.tea.2019.0278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Three-dimensional (3D)-engineered scaffolds have been widely investigated as drug delivery systems (DDS) or cancer models with the aim to develop effective cancer therapies. The in vitro and in vivo models developed via 3D printing (3DP) and tissue engineering concepts have significantly contributed to our understanding of cell-cell and cell-extracellular matrix interactions in the cancer microenvironment. Moreover, 3D tumor models were used to study the therapeutic efficiency of anticancer drugs. The present study aims to provide an overview of applying the 3DP and tissue engineering concepts for cancer studies with suggestions for future research directions. The 3DP technologies being used for the fabrication of personalized DDS have been highlighted and the potential technical approaches and challenges associated with the fused deposition modeling, the inkjet-powder bed, and stereolithography as the most promising 3DP techniques for drug delivery purposes are briefly described. Then, the advances, challenges, and future perspectives in tissue-engineered cancer models for precision medicine are discussed. Overall, future advances in this arena depend on the continuous integration of knowledge from cancer biology, biofabrication techniques, multiomics and patient data, and medical needs to develop effective treatments ultimately leading to improved clinical outcomes. Impact statement Three-dimensional printing (3DP) enables the fabrication of personalized medicines and drug delivery systems. The convergence of 3DP, tissue engineering concepts, and cancer biology could significantly improve our understanding of cancer biology and contribute to the development of new cancer therapies.
Collapse
Affiliation(s)
- Abbas Shafiee
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia.,Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia.,Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| |
Collapse
|
175
|
Miller CP, Shin W, Ahn EH, Kim HJ, Kim DH. Engineering Microphysiological Immune System Responses on Chips. Trends Biotechnol 2020; 38:857-872. [PMID: 32673588 DOI: 10.1016/j.tibtech.2020.01.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Tissues- and organs-on-chips are microphysiological systems (MPSs) that model the architectural and functional complexity of human tissues and organs that is lacking in conventional cell monolayer cultures. While substantial progress has been made in a variety of tissues and organs, chips recapitulating immune responses have not advanced as rapidly. This review discusses recent progress in MPSs for the investigation of immune responses. To illustrate recent developments, we focus on two cases in point: immunocompetent tumor microenvironment-on-a-chip devices that incorporate stromal and immune cell components and pathomimetic modeling of human mucosal immunity and inflammatory crosstalk. More broadly, we discuss the development of systems immunology-on-a-chip devices that integrate microfluidic engineering approaches with high-throughput omics measurements and emerging immunological applications of MPSs.
Collapse
Affiliation(s)
- Chris P Miller
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, 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; Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
176
|
Altay G, Batlle E, Fernández-Majada V, Martinez E. In vitro Self-organized Mouse Small Intestinal Epithelial Monolayer Protocol. Bio Protoc 2020; 10:e3514. [PMID: 33654739 DOI: 10.21769/bioprotoc.3514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/16/2019] [Accepted: 12/29/2019] [Indexed: 11/02/2022] Open
Abstract
Developing protocols to obtain intestinal epithelial monolayers that recapitulate in vivo physiology to overcome the limitations of the organoids' closed geometry has become of great interest during the last few years. Most of the developed culture models showed physiological-relevant cell composition but did not prove self-renewing capacities. Here, we show a simple method to obtain mouse small intestine-derived epithelial monolayers organized into proliferative crypt-like domains, containing stem cells, and differentiated villus-like regions, closely resembling the in vivo cell composition and distribution. In addition, we adapted our model to a tissue culture format compatible with functional studies and prove close to physiological barrier properties of our in vitro epithelial monolayers. Thus, we have set-up a protocol to generate physiologically relevant intestinal epithelial monolayers to be employed in assays where independent access to both luminal and basolateral compartments is needed, such as drug absorption, intracellular trafficking and microbiome-epithelium interaction assays.
Collapse
Affiliation(s)
- Gizem Altay
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Eduard Batlle
- Colorectal Cancer Laboratory, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10-12, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.,ICREA, Passeig Lluís Companys 23, 08010 Barcelona, Spain
| | - Vanesa Fernández-Majada
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elena Martinez
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, 08028 Spain
| |
Collapse
|
177
|
Snyder J, Wang CM, Zhang AQ, Li Y, Luchan J, Hosic S, Koppes R, Carrier RL, Koppes A. Materials and Microenvironments for Engineering the Intestinal Epithelium. Ann Biomed Eng 2020; 48:1916-1940. [DOI: 10.1007/s10439-020-02470-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
|
178
|
Suzuki A, Kato H, Kawakami T, Kodama Y, Shiozawa M, Kuwae H, Miwa K, Hoshikawa E, Haga K, Shiomi A, Uenoyama A, Saitoh I, Hayasaki H, Mizuno J, Izumi K. Development of microstructured fish scale collagen scaffolds to manufacture a tissue-engineered oral mucosa equivalent. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:578-600. [DOI: 10.1080/09205063.2019.1706147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ayako Suzuki
- Division of Biomimetics, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
- Division of Pediatric Dentistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroko Kato
- Division of Biomimetics, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | | | | | - Mayuko Shiozawa
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Hiroyuki Kuwae
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Keito Miwa
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Emi Hoshikawa
- Division of Biomimetics, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | - Kenta Haga
- Division of Biomimetics, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | - Aki Shiomi
- Division of Dental Education Research Development, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Atsushi Uenoyama
- Division of Oral and Maxillofacial Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Issei Saitoh
- Division of Pediatric Dentistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Haruaki Hayasaki
- Division of Pediatric Dentistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun Mizuno
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Kenji Izumi
- Division of Biomimetics, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| |
Collapse
|
179
|
Hinman SS, Kim R, Wang Y, Phillips KS, Attayek PJ, Allbritton NL. Microphysiological System Design: Simplicity Is Elegance. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 13:94-102. [PMID: 32095672 DOI: 10.1016/j.cobme.2019.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Design parameters for microphysiological systems (MPS) are driven by the need for new tools to answer questions focusing on human physiology in a robust and reliable manner. Within this perspective, engineering benchmarks and principles are identified to guide the construction of new devices in the MPS field, with emphasis placed on the design principles common to all tissues, as well as those unique to a subset of tissues. Leading organ replica technologies that recapitulate various functions of the brain, heart, intestine, and lung are highlighted as examples that meet the identified benchmarks and standards, with current barriers for large scale production and commercialization discussed. To reach their full potential and achieve widespread use, MPS will have to be recognized officially by government agencies, and toward this end, considerations of MPS as a potential regulatory tool are presented.
Collapse
Affiliation(s)
- Samuel S Hinman
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Raehyun Kim
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, and North Carolina State University, Raleigh, NC 27607, USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - K Scott Phillips
- United States Food and Drug Administration, Office of Medical Products and Tobacco, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Biology, Chemistry and Materials Science, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, USA
| | - Peter J Attayek
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, and North Carolina State University, Raleigh, NC 27607, USA
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, and North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
180
|
Xiang Y, Wen H, Yu Y, Li M, Fu X, Huang S. Gut-on-chip: Recreating human intestine in vitro. J Tissue Eng 2020; 11:2041731420965318. [PMID: 33282173 PMCID: PMC7682210 DOI: 10.1177/2041731420965318] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023] Open
Abstract
The human gut is important for food digestion and absorption, as well as a venue for a large number of microorganisms that coexist with the host. Although numerous in vitro models have been proposed to study intestinal pathology or interactions between intestinal microbes and host, they are far from recapitulating the real intestinal microenvironment in vivo. To assist researchers in further understanding gut physiology, the intestinal microbiome, and disease processes, a novel technology primarily based on microfluidics and cell biology, called "gut-on-chip," was developed to simulate the structure, function, and microenvironment of the human gut. In this review, we first introduce various types of gut-on-chip systems, then highlight their applications in drug pharmacokinetics, host-gut microbiota crosstalk, and nutrition metabolism. Finally, we discuss challenges in this field and prospects for better understanding interactions between intestinal flora and human hosts, and then provide guidance for clinical treatment of related diseases.
Collapse
Affiliation(s)
- Yunqing Xiang
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Wen
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yue Yu
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiongfei Fu
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuqiang Huang
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
181
|
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
|
182
|
Liu H, Wang Y, Cui K, Guo Y, Zhang X, Qin J. Advances in Hydrogels in Organoids and Organs-on-a-Chip. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902042. [PMID: 31282047 DOI: 10.1002/adma.201902042] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/25/2019] [Indexed: 05/10/2023]
Abstract
Significant advances in materials, microscale technology, and stem cell biology have enabled the construction of 3D tissues and organs, which will ultimately lead to more effective diagnostics and therapy. Organoids and organs-on-a-chip (OOC), evolved from developmental biology and bioengineering principles, have emerged as major technological breakthrough and distinct model systems to revolutionize biomedical research and drug discovery by recapitulating the key structural and functional complexity of human organs in vitro. There is growing interest in the development of functional biomaterials, especially hydrogels, for utilization in these promising systems to build more physiologically relevant 3D tissues with defined properties. The remarkable properties of defined hydrogels as proper extracellular matrix that can instruct cellular behaviors are presented. The recent trend where functional hydrogels are integrated into organoids and OOC systems for the construction of 3D tissue models is highlighted. Future opportunities and perspectives in the development of advanced hydrogels toward accelerating organoids and OOC research in biomedical applications are also discussed.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kangli Cui
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
183
|
De Gregorio V, Corrado B, Sbrescia S, Sibilio S, Urciuolo F, Netti PA, Imparato G. Intestine-on-chip device increases ECM remodeling inducing faster epithelial cell differentiation. Biotechnol Bioeng 2019; 117:556-566. [PMID: 31598957 DOI: 10.1002/bit.27186] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/11/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022]
Abstract
An intestine-on-chip has been developed to study intestinal physiology and pathophysiology as well as intestinal transport absorption and toxicity studies in a controlled and human similar environment. Here, we report that dynamic culture of an intestine-on-chip enhances extracellular matrix (ECM) remodeling of the stroma, basement membrane production and speeds up epithelial differentiation. We developed a three-dimensional human intestinal stromal equivalent composed of human intestinal subepithelial myofibroblasts embedded in their own ECM. Then, we cultured human colon carcinoma-derived cells in both static and dynamic conditions in the opportunely designed microfluidic system until the formation of a well-oriented epithelium. This low cost and handy microfluidic device allows to qualitatively and quantitatively detect epithelial polarization and mucus production as well as monitor barrier function and ECM remodeling after nutraceutical treatment.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - Brunella Corrado
- Departments of Naples, National Research Council, Institute for Microelectronics and Microsystems, Naples, Italy
| | | | - Sara Sibilio
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
| | - Francesco Urciuolo
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy.,Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy.,Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy.,Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| |
Collapse
|
184
|
Hinman SS, Wang Y, Allbritton NL. Photopatterned Membranes and Chemical Gradients Enable Scalable Phenotypic Organization of Primary Human Colon Epithelial Models. Anal Chem 2019; 91:15240-15247. [PMID: 31692334 DOI: 10.1021/acs.analchem.9b04217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Biochemical gradients across the intestinal epithelium play a major role in governing intestinal stem cell compartmentalization, differentiation dynamics, and organ-level self-renewal. However, scalable platforms that recapitulate the architecture and gradients present in vivo are absent. We present a platform in which individually addressable arrays of chemical gradients along the intestinal crypt long axis can be generated, enabling scalable culture of primary in vitro colonic epithelial replicas. The platform utilizes standardized well plate spacing, maintains access to basal and luminal compartments, and relies on a photopatterned porous membrane to act as diffusion windows while supporting the in vitro crypts. Simultaneous fabrication of 3875 crypts over a single membrane was developed. Growth factor gradients were modeled and then experimentally optimized to promote long-term health and self-renewal of the crypts which were assayed in situ by confocal fluorescence microscopy. The cultured in vitro crypt arrays successfully recapitulated the architecture and luminal-to-basal phenotypic polarity observed in vivo. Furthermore, known signaling regulators (e.g., butyrate and DAPT) produced measurable and predictable effects on the organized cell compartments, each decreasing crypt proliferation in the basal regions to negligible values. This platform is readily adaptable to the screening of tissue from individual patients to assay the impact of food and bacterial metabolites and/or drugs on colonic crypt dynamics. Importantly, the cassette is compatible with a wide range of sensing/detection modalities, and the developed fabrication methods should find applications for other cell and tissue types.
Collapse
Affiliation(s)
- Samuel S Hinman
- Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Yuli Wang
- Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Nancy L Allbritton
- Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States.,Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States , and North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
185
|
Pahapale GJ, Gao S, Romer LH, Gracias DH. Hierarchically Curved Gelatin for 3D Biomimetic Cell Culture. ACS APPLIED BIO MATERIALS 2019; 2:6004-6011. [DOI: 10.1021/acsabm.9b00916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
186
|
Speer JE, Wang Y, Fallon JK, Smith PC, Allbritton NL. Evaluation of human primary intestinal monolayers for drug metabolizing capabilities. J Biol Eng 2019; 13:82. [PMID: 31709009 PMCID: PMC6829970 DOI: 10.1186/s13036-019-0212-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The intestinal epithelium is a major site of drug metabolism in the human body, possessing enterocytes that house brush border enzymes and phase I and II drug metabolizing enzymes (DMEs). The enterocytes are supported by a porous extracellular matrix (ECM) that enables proper cell adhesion and function of brush border enzymes, such as alkaline phosphatase (ALP) and alanyl aminopeptidase (AAP), phase I DMEs that convert a parent drug to a more polar metabolite by introducing or unmasking a functional group, and phase II DMEs that form a covalent conjugate between a functional group on the parent compound or sequential metabolism of phase I metabolite. In our effort to develop an in vitro intestinal epithelium model, we investigate the impact of two previously described simple and customizable scaffolding systems, a gradient cross-linked scaffold and a conventional scaffold, on the ability of intestinal epithelial cells to produce drug metabolizing proteins as well as to metabolize exogenously added compounds. While the scaffolding systems possess a range of differences, they are most distinguished by their stiffness with the gradient cross-linked scaffold possessing a stiffness similar to that found in the in vivo intestine, while the conventional scaffold possesses a stiffness several orders of magnitude greater than that found in vivo. RESULTS The monolayers on the gradient cross-linked scaffold expressed CYP3A4, UGTs 2B17, 1A1 and 1A10, and CES2 proteins at a level similar to that in fresh crypts/villi. The monolayers on the conventional scaffold expressed similar levels of CYP3A4 and UGTs 1A1 and 1A10 DMEs to that found in fresh crypts/villi but significantly decreased expression of UGT2B17 and CES2 proteins. The activity of CYP3A4 and UGTs 1A1 and 1A10 was inducible in cells on the gradient cross-linked scaffold when the cells were treated with known inducers, whereas the CYP3A4 and UGT activities were not inducible in cells grown on the conventional scaffold. Both monolayers demonstrate esterase activity but the activity measured in cells on the conventional scaffold could not be inhibited with a known CES2 inhibitor. Both monolayer culture systems displayed similar ALP and AAP brush border enzyme activity. When cells on the conventional scaffold were incubated with a yes-associated protein (YAP) inhibitor, CYP3A4 activity was greatly enhanced suggesting that mechano-transduction signaling can modulate drug metabolizing enzymes. CONCLUSIONS The use of a cross-linked hydrogel scaffold for expansion and differentiation of primary human intestinal stem cells dramatically impacts the induction of CYP3A4 and maintenance of UGT and CES drug metabolizing enzymes in vitro making this a superior substrate for enterocyte culture in DME studies. This work highlights the influence of mechanical properties of the culture substrate on protein expression and the activity of drug metabolizing enzymes as a critical factor in developing accurate assay protocols for pharmacokinetic studies using primary intestinal cells. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Jennifer E. Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607 USA
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607 USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607 USA
| |
Collapse
|
187
|
Development of a villi-like micropatterned porous membrane for intestinal magnesium and calcium uptake studies. Acta Biomater 2019; 99:110-120. [PMID: 31465881 DOI: 10.1016/j.actbio.2019.08.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 12/25/2022]
Abstract
Intestinal enterocytes are key players in the absorption of magnesium (Mg2+) and calcium (Ca2+). Understanding the exact molecular mechanisms by which their absorption behavior is regulated could greatly improve treatment strategies for stimulating intestinal absorption in diseases with Mg2+ and/or Ca2+ deficiency. However, such studies are hampered by the lack of in vitro intestinal cell models mimicking the mechanical and physiological properties of the gut. In this study we develop an in vitro gut model based on porous micropatterned membranes with villi-like surface topography and mechanical properties closely mimicking that of intestinal tissue. These membranes are prepared via phase separation micromolding using poly-ε-caprolactone/poly-lactic-glycolic acid (PCL/PLGA) polymer blend and can facilitate cellular differentiation of Caco-2 cells similar to native enterocytes. In fact, cells cultured on these micropatterned membranes form a brush border of microvilli with spatial differences in morphology and tight junction formation along the villous-base axis. Moreover, cells cultured on our membranes show a 2-fold increased alkaline phosphatase activity at the end of differentiation. Finally, we demonstrate that cells cultured on our micropatterned membranes have a 4- and 1.5-fold increased uptake of 25Mg and 45Ca, respectively, compared to non-patterned membranes. These results indicate that the new membranes can mimic the intestinal environment and therefore can have a great impact on mineral uptake in vitro. STATEMENT OF SIGNIFICANCE: This study presents the development of an in vitro gut model consisting of villi-like PCL/PLGA micropatterned membranes. These membranes are prepared via phase separation micromolding (PSμM), a technique which allows tailoring of the membrane surface topography combined with membrane porosity and interconnectivity which are important parameters for membranes used for in vitro transport studies. The culture of Caco-2 cells on these micropatterned membranes shows that they facilitate cellular differentiation similar to gut enterocytes. Our data indicate that mimicking the 3D geometry of the gut is very important for improving the physiological relevance of in vitro gut models. In the future, our micropatterned membranes with segment-specific geometries, in combination with isotopic measurements, would be applied to perform detailed ion uptake and transport studies.
Collapse
|
188
|
Ladd MR, Costello CM, Gosztyla C, Werts AD, Johnson B, Fulton WB, Martin LY, Redfield EJ, Crawford B, Panaparambil R, Sodhi CP, March JC, Hackam DJ. Development of Intestinal Scaffolds that Mimic Native Mammalian Intestinal Tissue. Tissue Eng Part A 2019; 25:1225-1241. [PMID: 30652526 PMCID: PMC6760185 DOI: 10.1089/ten.tea.2018.0239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/05/2018] [Indexed: 12/27/2022] Open
Abstract
IMPACT STATEMENT This study is significant because it demonstrates an attempt to design a scaffold specifically for small intestine using a novel fabrication method, resulting in an architecture that resembles intestinal villi. In addition, we use the versatile polymer poly(glycerol sebacate) (PGS) for artificial intestine, which has tunable mechanical and degradation properties that can be harnessed for further fine-tuning of scaffold design. Moreover, the utilization of PGS allows for future development of growth factor and drug delivery from the scaffolds to promote artificial intestine formation.
Collapse
Affiliation(s)
- Mitchell R. Ladd
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Cait M. Costello
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York
| | - Carolyn Gosztyla
- Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Adam D. Werts
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Blake Johnson
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - William B. Fulton
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Laura Y. Martin
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Elizabeth J. Redfield
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York
| | - Bryan Crawford
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Rohan Panaparambil
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Chhinder P. Sodhi
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John C. March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York
| | - David J. Hackam
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
189
|
Luu L, Johnston LJ, Derricott H, Armstrong SD, Randle N, Hartley CS, Duckworth CA, Campbell BJ, Wastling JM, Coombes JL. An Open-Format Enteroid Culture System for Interrogation of Interactions Between Toxoplasma gondii and the Intestinal Epithelium. Front Cell Infect Microbiol 2019; 9:300. [PMID: 31555604 PMCID: PMC6723115 DOI: 10.3389/fcimb.2019.00300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022] Open
Abstract
When transmitted through the oral route, Toxoplasma gondii first interacts with its host at the small intestinal epithelium. This interaction is crucial to controlling initial invasion and replication, as well as shaping the quality of the systemic immune response. It is therefore an attractive target for the design of novel vaccines and adjuvants. However, due to a lack of tractable infection models, we understand surprisingly little about the molecular pathways that govern this interaction. The in vitro culture of small intestinal epithelium as 3D enteroids shows great promise for modeling the epithelial response to infection. However, the enclosed luminal space makes the application of infectious agents to the apical epithelial surface challenging. Here, we have developed three novel enteroid-based techniques for modeling T. gondii infection. In particular, we have adapted enteroid culture protocols to generate collagen-supported epithelial sheets with an exposed apical surface. These cultures retain epithelial polarization, and the presence of fully differentiated epithelial cell populations. They are susceptible to infection with, and support replication of, T. gondii. Using quantitative label-free mass spectrometry, we show that T. gondii infection of the enteroid epithelium is associated with up-regulation of proteins associated with cholesterol metabolism, extracellular exosomes, intermicrovillar adhesion, and cell junctions. Inhibition of host cholesterol and isoprenoid biosynthesis with Atorvastatin resulted in a reduction in parasite load only at higher doses, indicating that de novo synthesis may support, but is not required for, parasite replication. These novel models therefore offer tractable tools for investigating how interactions between T. gondii and the host intestinal epithelium influence the course of infection.
Collapse
Affiliation(s)
- Lisa Luu
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Luke J. Johnston
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Hayley Derricott
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Stuart D. Armstrong
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Nadine Randle
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Catherine S. Hartley
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Carrie A. Duckworth
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Barry J. Campbell
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Jonathan M. Wastling
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Janine L. Coombes
- Department of Infection Biology, Faculty of Health and Life Sciences, School of Veterinary Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
190
|
Current understanding of the gut microbiota shaping mechanisms. J Biomed Sci 2019; 26:59. [PMID: 31434568 PMCID: PMC6702754 DOI: 10.1186/s12929-019-0554-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Increasing evidences have shown strong associations between gut microbiota and many human diseases, and understanding the dynamic crosstalks of host-microbe interaction in the gut has become necessary for the detection, prevention, or therapy of diseases. Many reports have showed that diet, nutrient, pharmacologic factors and many other stimuli play dominant roles in the modulation of gut microbial compositions. However, it is inappropriate to neglect the impact of host factors on shaping the gut microbiota. In this review, we highlighted the current findings of the host factors that could modulate the gut microbiota. Particularly the epithelium-associated factors, including the innate immune sensors, anti-microbial peptides, mucus barrier, secretory IgAs, epithelial microvilli, epithelial tight junctions, epithelium metabolism, oxygen barrier, and even the microRNAs are discussed in the context of the microbiota shaping. With these shaping factors, the gut epithelial cells could select the residing microbes and affect the microbial composition. This knowledge not only could provide the opportunities to better control many diseases, but may also be used for predicting the success of fecal microbiota transplantation clinically.
Collapse
|
191
|
Creff J, Courson R, Mangeat T, Foncy J, Souleille S, Thibault C, Besson A, Malaquin L. Fabrication of 3D scaffolds reproducing intestinal epithelium topography by high-resolution 3D stereolithography. Biomaterials 2019; 221:119404. [PMID: 31419651 DOI: 10.1016/j.biomaterials.2019.119404] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/26/2019] [Accepted: 08/01/2019] [Indexed: 12/20/2022]
Abstract
The small intestine is a complex tissue with a crypt/villus architecture and high tissue polarity. Maintenance of tissue integrity and function is supported by a constant renewal of the epithelium, with proliferative cells located in the crypts and differentiated cells migrating upward to the top of villi. So far, most in vitro studies have been limited to 2D surfaces or 3D organoid cultures that do not fully recapitulate the tissue 3D architecture, microenvironment and cell compartmentalization found in vivo. Here, we report the development of a 3D model that reproduces more faithfully the architecture of the intestinal epithelium in vitro. We developed a new fabrication process combining a photopolymerizable hydrogel that supports the growth of intestinal cell lines with high-resolution stereolithography 3D printing. This approach offers the possibility to create artificial 3D scaffolds matching the dimensions and architecture of mouse intestinal crypts and villi. We demonstrate that these 3D culture models support the growth and differentiation of Caco-2 cells for 3 weeks. These models may constitute a complementary approach to organoid cultures to study intestinal homeostasis by allowing guided self-organization and controlled differentiation, as well as for in vitro drug screening and testing.
Collapse
Affiliation(s)
- Justine Creff
- LBCMCP, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, 31062, Toulouse Cedex, France; LAAS-CNRS, Université de Toulouse, CNRS, F-31400, Toulouse, France
| | - Rémi Courson
- LAAS-CNRS, Université de Toulouse, CNRS, F-31400, Toulouse, France
| | - Thomas Mangeat
- LBCMCP, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, 31062, Toulouse Cedex, France
| | - Julie Foncy
- LAAS-CNRS, Université de Toulouse, CNRS, F-31400, Toulouse, France
| | | | - C Thibault
- LAAS-CNRS, Université de Toulouse, CNRS, F-31400, Toulouse, France; Université de Toulouse, Institut National des Sciences Appliquées-INSA, F-31400 Toulouse, France
| | - Arnaud Besson
- LBCMCP, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, 31062, Toulouse Cedex, France.
| | - Laurent Malaquin
- LAAS-CNRS, Université de Toulouse, CNRS, F-31400, Toulouse, France.
| |
Collapse
|
192
|
Brovold M, Almeida JI, Pla-Palacín I, Sainz-Arnal P, Sánchez-Romero N, Rivas JJ, Almeida H, Dachary PR, Serrano-Aulló T, Soker S, Baptista PM. Naturally-Derived Biomaterials for Tissue Engineering Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1077:421-449. [PMID: 30357702 PMCID: PMC7526297 DOI: 10.1007/978-981-13-0947-2_23] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Naturally-derived biomaterials have been used for decades in multiple regenerative medicine applications. From the simplest cell microcarriers made of collagen or alginate, to highly complex decellularized whole-organ scaffolds, these biomaterials represent a class of substances that is usually first in choice at the time of electing a functional and useful biomaterial. Hence, in this chapter we describe the several naturally-derived biomaterials used in tissue engineering applications and their classification, based on composition. We will also describe some of the present uses of the generated tissues like drug discovery, developmental biology, bioprinting and transplantation.
Collapse
Affiliation(s)
- Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Joana I Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Iris Pla-Palacín
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Pilar Sainz-Arnal
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
- Aragon Health Sciences Institute (IACS), Zaragoza, Spain
| | | | - Jesus J Rivas
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Helen Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Pablo Royo Dachary
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Liver Transplant Unit, Gastroenterology Department, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Trinidad Serrano-Aulló
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Liver Transplant Unit, Gastroenterology Department, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA.
| | - Pedro M Baptista
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain.
- Center for Biomedical Research Network Liver and Digestive Diseases (CIBERehd), Zaragoza, Spain.
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.
- Biomedical and Aerospace Engineering Department, Universidad Carlos III de Madrid, Madrid, Spain.
- Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
193
|
Wang Y, Shao Z, Zheng W, Xie Y, Luo G, Ding M, Liang Q. A 3D construct of the intestinal canal with wrinkle morphology on a centrifugation configuring microfluidic chip. Biofabrication 2019; 11:045001. [DOI: 10.1088/1758-5090/ab21b0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
194
|
Astragalus polysaccharides attenuated inflammation and balanced the gut microflora in mice challenged with Salmonella typhimurium. Int Immunopharmacol 2019; 74:105681. [PMID: 31220694 DOI: 10.1016/j.intimp.2019.105681] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 01/25/2023]
Abstract
Salmonella typhimurium (S. t.) is one of the main pathogens that causes acute gastroenteritis. To evaluate the anti-inflammatory mechanism of Astragalus polysaccharide (APS) in vivo and its influence on the intestinal flora, BALB/c mice were infected with S. t. to establish a model of diarrhea. The disease activity index (DAI) scores showed that APS attenuated S. t.-induced weight loss and diarrhea in mice. APS significantly reduced the index of the liver and spleen as well as the ALT and AST levels in serum (P < 0.05). Hematoxylin and eosin (H&E) results indicated that APS significantly increased jejunum villus height and crypt depth and reduced the infiltration of inflammatory cells (P < 0.05). Additionally, APS increased the tight junction (TJ) proteins expression levels of ZO-1, Occludin and Claudin-1 in the jejunum. The results of 16S rDNA showed that APS significantly increased the number of Lactobacillus and Bifidobacterium spp. to normal levels (compared with the control group). In addition, APS significantly decreased the mRNA expression levels of the proinflammatory cytokines TNF-α, IL-1β, IL-6 and IL-17 in the jejunum (P < 0.01) as well as the proteins expression levels of COX-2 and iNOS (P < 0.05). Western blot confirmed that prefeeding with APS inhibited S. t.-induced expression of TLR4 and MyD88 in the jejunum and further inhibited nuclear factor-κB (NF-κB) activation, including the nuclear translocation of the p65 NF-κB subunit and the phosphorylation and degradation of IκB-α. This was the key to APS inhibition of the production of inflammatory factors and inflammatory mediators in the jejunum.
Collapse
|
195
|
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
|
196
|
Abstract
Recent studies have demonstrated an array of stem cell-derived, self-organizing miniature organs, termed organoids, that replicate the key structural and functional characteristics of their in vivo counterparts. As organoid technology opens up new frontiers of research in biomedicine, there is an emerging need for innovative engineering approaches for the production, control, and analysis of organoids and their microenvironment. In this Review, we explore organ-on-a-chip technology as a platform to fulfill this need and examine how this technology may be leveraged to address major technical challenges in organoid research. We also discuss emerging opportunities and future obstacles for the development and application of organoid-on-a-chip technology.
Collapse
Affiliation(s)
- Sunghee Estelle Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrei Georgescu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
197
|
Huang J, Ren Y, Wu X, Li Z, Ren J. Gut bioengineering promotes gut repair and pharmaceutical research: a review. J Tissue Eng 2019; 10:2041731419839846. [PMID: 31037215 PMCID: PMC6475831 DOI: 10.1177/2041731419839846] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract has a diverse set of physiological functions, including peristalsis, immune defense, and nutrient absorptions. These functions are mediated by various intestinal cells such as epithelial cells, interstitial cells, smooth muscle cells, and neurocytes. The loss or dysfunction of specific cells directly results in GI disease, while supplementation of normal cells promotes gut healing. Gut bioengineering has been developing for this purpose to reconstruct the damaged tissues. Moreover, GI tract provides an accessible route for drug delivery, but the collateral damages induced by side effects cannot be ignored. Bioengineered intestinal tissues provide three-dimensional platforms that mimic the in vivo environment to study drug functions. Given the importance of gut bioengineering in current research, in this review, we summarize the advances in the technologies of gut bioengineering and their applications. We were able to identify several ground-breaking discoveries in our review, while more work is needed to promote the clinical translation of gut bioengineering.
Collapse
Affiliation(s)
- Jinjian Huang
- School of Medicine, Southeast University, Nanjing, China.,Laboratory for Trauma and Surgical Infections, Department of Surgery, Jinling Hospital, Nanjing, China
| | - Yanhan Ren
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Xiuwen Wu
- Laboratory for Trauma and Surgical Infections, Department of Surgery, Jinling Hospital, Nanjing, China
| | - Zongan Li
- School of NARI Electrical and Automation Engineering, Nanjing Normal University, Nanjing, China
| | - Jianan Ren
- School of Medicine, Southeast University, Nanjing, China.,Laboratory for Trauma and Surgical Infections, Department of Surgery, Jinling Hospital, Nanjing, China
| |
Collapse
|
198
|
Holloway EM, Capeling MM, Spence JR. Biologically inspired approaches to enhance human organoid complexity. Development 2019; 146:dev166173. [PMID: 30992275 PMCID: PMC6503984 DOI: 10.1242/dev.166173] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organoids are complex three-dimensional in vitro organ-like model systems. Human organoids, which are derived from human pluripotent stem cells or primary human donor tissue, have been used to address fundamental questions about human development, stem cell biology and organ regeneration. Focus has now shifted towards implementation of organoids for biological discovery and advancing existing systems to more faithfully recapitulate the native organ. This work has highlighted significant unknowns in human biology and has invigorated new exploration into the cellular makeup of human organs during development and in the adult - work that is crucial for providing appropriate benchmarks for organoid systems. In this Review, we discuss efforts to characterize human organ cellular complexity and attempts to make organoid models more realistic through co-culture, transplantation and bioengineering approaches.
Collapse
Affiliation(s)
- Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
199
|
O'Driscoll CM, Bernkop-Schnürch A, Friedl JD, Préat V, Jannin V. Oral delivery of non-viral nucleic acid-based therapeutics - do we have the guts for this? Eur J Pharm Sci 2019; 133:190-204. [PMID: 30946964 DOI: 10.1016/j.ejps.2019.03.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/22/2022]
Abstract
Gene therapy with RNA and pDNA-based drugs is limited by poor enzymatic stability and poor cellular permeation. The delivery of nucleic acids, in particular by the oral route, remains a major hurdle. This review will focus on the barriers to the oral delivery of nucleic acids and the strategies, in particular formulation strategies, which have been developed to overcome these barriers. Due to their very low oral bioavailability, the most obvious and most investigated biomedical applications for their oral delivery are related to the local treatment of inflammatory bowel diseases and colorectal cancers. Preclinical data but not yet clinical studies support the potential use of the oral route for the local delivery of formulated nucleic acid-based drugs.
Collapse
Affiliation(s)
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Julian D Friedl
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Véronique Préat
- Universite catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1.73.12, 1200 Brussels, Belgium.
| | - Vincent Jannin
- Gattefossé SAS, 36 chemin de Genas, 69804 Saint-Priest cedex, France.
| |
Collapse
|
200
|
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
|