1
|
Sibilio S, Mennella R, Gregorio VD, Rocca AL, Urciuolo F, Imparato G, Netti PA. A novel membrane-on-chip guides morphogenesis for the reconstruction of the intestinal crypt-villus axis. Biofabrication 2024; 16:045019. [PMID: 39029501 DOI: 10.1088/1758-5090/ad6599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/19/2024] [Indexed: 07/21/2024]
Abstract
Reconstructing the microscale villous organisation and functionality of the small intestine is essential for developingin vitroplatforms tailored for absorption studies as well as for investigating intestinal morphogenesis in development and disease. However, the current fabrication techniques able to mimic the villus-crypt axis poses significant challenges in terms of reconstruction of the complex 3D microarchitecture. These challenges extend beyond mere structural intricacies to encompass the incorporation of diverse cell types and the management of intricate fluid dynamics within the system. Here, we introduce a novel microfluidic device calledIn-Crypts, which integrates a cell-instructive membrane aimed at inducing and guiding Caco-2 cells morphogenesis. Patterned topographical cues embossed onto the porous membrane induce the formation of a well-organized intestinal epithelium, characterized by proliferating crypt-like domains and differentiated villus-like regions. Notably, our cell-instructive porous membrane effectively sustains stem cells development, faithfully replicating the niche environment ofin vivointestinal crypts thus mirroring the cell biogeography observedin vivo. Moreover, by introducing dynamic fluid flow, we provide a faithful recapitulation of the native microenvironmental shear stress experienced by the intestinal epithelium. This stress plays a crucial role in influencing cell behaviour, differentiation, and overall functionality, thus offering a highly realistic model for studying intestinal physiology and pathology. The resulting intestinal epithelium exhibits significantly denser regions of mucus and microvilli, characteristic typically absent in static cultures, upregulating more than 1.5 of the amount expressed in the classical flattened configuration, enhanced epithelial cell differentiation and increased adsorptive surface area. Hence, the innovative design ofIn-Cryptsproves the critical role of employing a cell-instructive membrane in argument the physiological relevance of organs-on-chips. This aspect, among others, will contribute to a more comprehensive understanding of organism function, directly impacting drug discovery and development.
Collapse
Affiliation(s)
- Sara Sibilio
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Naples, Italy
| | - Raffaele Mennella
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Naples, Italy
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Naples, Italy
| | - Vincenza De Gregorio
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Naples, Italy
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Alessia La Rocca
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Naples, Italy
| | - Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Naples, Italy
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Naples, Italy
| | - Giorgia Imparato
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Naples, Italy
| | - Paolo A Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Naples, Italy
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Naples, Italy
| |
Collapse
|
2
|
Urciuolo F, Imparato G, Netti PA. Engineering Cell Instructive Microenvironments for In Vitro Replication of Functional Barrier Organs. Adv Healthc Mater 2024; 13:e2400357. [PMID: 38695274 DOI: 10.1002/adhm.202400357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/02/2024] [Indexed: 05/14/2024]
Abstract
Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
| | - Giorgia Imparato
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| |
Collapse
|
3
|
Wang H, Li X, Shi P, You X, Zhao G. Establishment and evaluation of on-chip intestinal barrier biosystems based on microfluidic techniques. Mater Today Bio 2024; 26:101079. [PMID: 38774450 PMCID: PMC11107260 DOI: 10.1016/j.mtbio.2024.101079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
As a booming engineering technology, the microfluidic chip has been widely applied for replicating the complexity of human intestinal micro-physiological ecosystems in vitro. Biosensors, 3D imaging, and multi-omics have been applied to engineer more sophisticated intestinal barrier-on-chip platforms, allowing the improved monitoring of physiological processes and enhancing chip performance. In this review, we report cutting-edge advances in the microfluidic techniques applied for the establishment and evaluation of intestinal barrier platforms. We discuss different design principles and microfabrication strategies for the establishment of microfluidic gut barrier models in vitro. Further, we comprehensively cover the complex cell types (e.g., epithelium, intestinal organoids, endothelium, microbes, and immune cells) and controllable extracellular microenvironment parameters (e.g., oxygen gradient, peristalsis, bioflow, and gut-organ axis) used to recapitulate the main structural and functional complexity of gut barriers. We also present the current multidisciplinary technologies and indicators used for evaluating the morphological structure and barrier integrity of established gut barrier models in vitro. Finally, we highlight the challenges and future perspectives for accelerating the broader applications of these platforms in disease simulation, drug development, and personalized medicine. Hence, this review provides a comprehensive guide for the development and evaluation of microfluidic-based gut barrier platforms.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Pengcheng Shi
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- CAS-Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
4
|
Liu T, Gu J, Fu C, Su L. Three-Dimensional Scaffolds for Intestinal Cell Culture: Fabrication, Utilization, and Prospects. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:158-175. [PMID: 37646409 DOI: 10.1089/ten.teb.2023.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The intestine is a visceral organ that integrates absorption, metabolism, and immunity, which is vulnerable to external stimulus. Researchers in the fields such as food science, immunology, and pharmacology have committed to developing appropriate in vitro intestinal cell models to study the intestinal absorption and metabolism mechanisms of various nutrients and drugs, or pathogenesis of intestinal diseases. In the past three decades, the intestinal cell models have undergone a significant transformation from conventional two-dimensional cultures to three-dimensional (3D) systems, and the achievements of 3D cell culture have been greatly contributed by the fabrication of different scaffolds. In this review, we first introduce the developing trend of existing intestinal models. Then, four types of scaffolds, including Transwell, hydrogel, tubular scaffolds, and intestine-on-a-chip, are discussed for their 3D structure, composition, advantages, and limitations in the establishment of intestinal cell models. Excitingly, some of the in vitro intestinal cell models based on these scaffolds could successfully mimic the 3D structure, microenvironment, mechanical peristalsis, fluid system, signaling gradients, or other important aspects of the original human intestine. Furthermore, we discuss the potential applications of the intestinal cell models in drug screening, disease modeling, and even regenerative repair of intestinal tissues. This review presents an overview of state-of-the-art scaffold-based cell models within the context of intestines, and highlights their major advances and applications contributing to a better knowledge of intestinal diseases. Impact statement The intestine tract is crucial in the absorption and metabolism of nutrients and drugs, as well as immune responses against external pathogens or antigens in a complex microenvironment. The appropriate experimental cell model in vitro is needed for in-depth studies of intestines, due to the limitation of animal models in dynamic control and real-time assessment of key intestinal physiological and pathological processes, as well as the "R" principles in laboratory animal experiments. Three-dimensional (3D) scaffold-based cell cultivation has become a developing tendency because of the superior cell proliferation and differentiation and more physiologically relevant environment supported by the customized 3D scaffolds. In this review, we summarize four types of up-to-date 3D cell culture scaffolds fabricated by various materials and techniques for a better recapitulation of some essential physiological and functional characteristics of original intestines compared to conventional cell models. These emerging 3D intestinal models have shown promising results in not only evaluating the pharmacokinetic characteristics, security, and effectiveness of drugs, but also studying the pathological mechanisms of intestinal diseases at cellular and molecular levels. Importantly, the weakness of the representative 3D models for intestines is also discussed.
Collapse
Affiliation(s)
- Tiange Liu
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Jia Gu
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Lingshan Su
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
5
|
Becker HM, Seidler UE. Bicarbonate secretion and acid/base sensing by the intestine. Pflugers Arch 2024; 476:593-610. [PMID: 38374228 PMCID: PMC11006743 DOI: 10.1007/s00424-024-02914-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/21/2024]
Abstract
The transport of bicarbonate across the enterocyte cell membrane regulates the intracellular as well as the luminal pH and is an essential part of directional fluid movement in the gut. Since the first description of "active" transport of HCO3- ions against a concentration gradient in the 1970s, the fundamental role of HCO3- transport for multiple intestinal functions has been recognized. The ion transport proteins have been identified and molecularly characterized, and knockout mouse models have given insight into their individual role in a variety of functions. This review describes the progress made in the last decade regarding novel techniques and new findings in the molecular regulation of intestinal HCO3- transport in the different segments of the gut. We discuss human diseases with defects in intestinal HCO3- secretion and potential treatment strategies to increase luminal alkalinity. In the last part of the review, the cellular and organismal mechanisms for acid/base sensing in the intestinal tract are highlighted.
Collapse
Affiliation(s)
- Holger M Becker
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany
| | - Ursula E Seidler
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
6
|
Jang Y, Jung J, Oh J. Bio-Microfabrication of 2D and 3D Biomimetic Gut-on-a-Chip. MICROMACHINES 2023; 14:1736. [PMID: 37763899 PMCID: PMC10537549 DOI: 10.3390/mi14091736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023]
Abstract
Traditional goal of microfabrication was to limitedly construct nano- and micro-geometries on silicon or quartz wafers using various semiconductor manufacturing technologies, such as photolithography, soft lithography, etching, deposition, and so on. However, recent integration with biotechnologies has led to a wide expansion of microfabrication. In particular, many researchers studying pharmacology and pathology are very interested in producing in vitro models that mimic the actual intestine to study the effectiveness of new drug testing and interactions between organs. Various bio-microfabrication techniques have been developed while solving inherent problems when developing in vitro micromodels that mimic the real large intestine. This intensive review introduces various bio-microfabrication techniques that have been used, until recently, to realize two-dimensional and three-dimensional biomimetic experimental models. Regarding the topic of gut chips, two major review subtopics and two-dimensional and three-dimensional gut chips were employed, focusing on the membrane-based manufacturing process for two-dimensional gut chips and the scaffold-based manufacturing process for three-dimensional gut chips, respectively.
Collapse
Affiliation(s)
- Yeongseok Jang
- Department of Mechanical Design Engineering, Jeonbuk National University, Jeonju-si 54896, Jeollabuk-do, Republic of Korea;
| | - Jinmu Jung
- Department of Nano-Bio Mechanical System Engineering, Jeonbuk National University, Jeonju-si 54896, Jeollabuk-do, Republic of Korea
| | - Jonghyun Oh
- Department of Nano-Bio Mechanical System Engineering, Jeonbuk National University, Jeonju-si 54896, Jeollabuk-do, Republic of Korea
| |
Collapse
|
7
|
Zhang M, Zuo Z, Zhang X, Wang L. Food biopolymer behaviors in the digestive tract: implications for nutrient delivery. Crit Rev Food Sci Nutr 2023; 64:8709-8727. [PMID: 37216487 DOI: 10.1080/10408398.2023.2202778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Biopolymers are prevalent in both natural and processed foods, serving as thickeners, emulsifiers, and stabilizers. Although specific biopolymers are known to affect digestion, the mechanisms behind their influence on the nutrient absorption and bioavailability in processed foods are not yet fully understood. The aim of this review is to elucidate the complex interplay between biopolymers and their behavior in vivo, and to provide insights into the possible physiological consequences of their consumption. The colloidization process of biopolymer in various phases of digestion was analyzed and its impact on nutrition absorption and gastrointestinal tract was summarized. Furthermore, the review discusses the methodologies used to assess colloidization and emphasizes the need for more realistic models to overcome challenges in practical applications. By controlling macronutrient bioavailability using biopolymers, it is possible to enhance health benefits, such as improving gut health, aiding in weight management, and regulating blood sugar levels. The physiological effect of extracted biopolymers utilized in modern food structuring technology cannot be predicted solely based on their inherent functionality. It is essential to account for factors such as their initial consuming state and interactions with other food components to better understand the potential health benefits of biopolymers.
Collapse
Affiliation(s)
- Ming Zhang
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhongyu Zuo
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xinxia Zhang
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Wang
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| |
Collapse
|
8
|
Rudolph SE, Longo BN, Tse MW, Houchin MR, Shokoufandeh MM, Chen Y, Kaplan DL. Crypt-Villus Scaffold Architecture for Bioengineering Functional Human Intestinal Epithelium. ACS Biomater Sci Eng 2022; 8:4942-4955. [PMID: 36191009 PMCID: PMC10379436 DOI: 10.1021/acsbiomaterials.2c00851] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Crypt-villus architecture in the small intestine is crucial for the structural integrity of the intestinal epithelium and maintenance of gut homeostasis. We utilized three-dimensional (3D) printing and inverse molding techniques to form three-dimensional (3D) spongy scaffold systems that resemble the intestinal crypt-villus microarchitecture. The scaffolds consist of silk fibroin protein with curved lumens with rows of protruding villi with invaginating crypts to generate the architecture. Intestinal cell (Caco-2, HT29-MTX) attachment and growth, as well as long-term culture support were demonstrated with cell polarization and tissue barrier properties compared to two-dimensional (2D) Transwell culture controls. Further, physiologically relevant oxygen gradients were generated in the 3D system. The various advantages of this system may be ascribed to the more physiologically relevant 3D environment, offering a system for the exploration of disease pathogenesis, host-microbiome interactions, and therapeutic discovery.
Collapse
Affiliation(s)
- Sara E Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Brooke N Longo
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Megan W Tse
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Megan R Houchin
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Mina M Shokoufandeh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
9
|
De Gregorio V, Sgambato C, Urciuolo F, Vecchione R, Netti PA, Imparato G. Immunoresponsive microbiota-gut-on-chip reproduces barrier dysfunction, stromal reshaping and probiotics translocation under inflammation. Biomaterials 2022; 286:121573. [PMID: 35617781 DOI: 10.1016/j.biomaterials.2022.121573] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 01/21/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022]
|
10
|
Luo Y, Liu Y, Shen Y, He J, Li H, Lan C, Li J, Chen H, Chen D, Ren Z, Yu B, Huang Z, Zheng P, Mao X, Yu J, Luo J, Yan H. Fermented Alfalfa Meal Instead of "Grain-Type" Feedstuffs in the Diet Improves Intestinal Health Related Indexes in Weaned Pigs. Front Microbiol 2021; 12:797875. [PMID: 34966376 PMCID: PMC8710769 DOI: 10.3389/fmicb.2021.797875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/15/2021] [Indexed: 01/14/2023] Open
Abstract
Corn and soybean meal are the two main components in formula feed of farm animals, leading to a serious food competition between humans and livestock. An alternative may be to encourage the utilization of unconventional feedstuff in animal diet. In the current study, we evaluated the utilization of fermented alfalfa meal (FAM) in weaned pigs. Twenty weaned piglets (separately caged) were randomly divided into two groups. Pigs in the control group (CON) were fed corn-soybean meal diet, and part of corn and soya protein concentrate in the diet of another group was replaced by 8% FAM. After 40 days of feeding, the average feed intake of FAM pigs was increased (P > 0.05), and the villus height (VH) of jejunum and duodenum, crypt depth (CD), and VH/CD in FAM pigs was improved compared to the CON group (P < 0.05). The increase (P < 0.05) of goblet cells in the jejunum of FAM pigs was positively correlated with the expression of MUC-2 gene (R = 0.9150). The expression of genes related to immunity (IRAK4, NF-κB, and IL-10) and intestinal barrier (Occludin and MUC-2) in the jejunum, as well as the expression of ZO-1 and MUC-2 in the colon of these pigs, also showed increase (P < 0.05) compared to CON pigs, which was accompanied by the decrease (P < 0.05) of LPS concentration in the serum. The elevated proportion of CD3+ and CD8+ T-lymphocyte subsets in spleen (P < 0.05) confirmed the improvement of systemic immune function in FAM pigs. In addition, FAM pigs have a higher β-diversity of microbial community (P < 0.05) and promoted enrichment of probiotics such as Lactobacillus that positively was correlated with acetate concentration in the colon over CON pigs. In summary, partially replacement of expanded corn and soya protein concentrate with FAM (8%) may benefit the intestinal barrier and immune function of weaned pigs without affecting their growth. Our findings also provide evidence of the feasibility of FAM as a dietary component in pigs to reduce the consumption of grain.
Collapse
Affiliation(s)
- Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Yang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Yuqing Shen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Hua Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Cong Lan
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jiayan Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Hong Chen
- College of Food Science, Sichuan Agricultural University, Ya’an, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Zhihua Ren
- Sichuan Province Key Laboratory of Animal Disease and Human Health, Key Laboratory of Environmental Hazard and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Hui Yan
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
11
|
Helena Macedo M, Baião A, Pinto S, Barros AS, Almeida H, Almeida A, das Neves J, Sarmento B. Mucus-producing 3D cell culture models. Adv Drug Deliv Rev 2021; 178:113993. [PMID: 34619286 DOI: 10.1016/j.addr.2021.113993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/23/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
In vitro cell-based models have been used for a long time since they are normally easily obtained and have an advantageous cost-benefit. Besides, they can serve a variety of ends, from studying drug absorption and metabolism to disease modeling. However, some in vitro models are too simplistic, not accurately representing the living tissues. It has been shown, mainly in the last years, that fully mimicking a tissue composition and architecture can be paramount for cellular behavior and, consequently, for the outcomes of the studies using such models. Because of this, 3D in vitro cell models have been gaining much attention, since they are able to better replicate the in vivo environment. In this review we focus on 3D models that contain mucus-producing cells, as mucus can play a pivotal role in drug absorption. Being frequently overlooked, this viscous fluid can have an impact on drug delivery. Thus, the aim of this review is to understand to which extent can mucus affect mucosal drug delivery and to provide a state-of-the-art report on the existing 3D cell-based mucus models.
Collapse
|
12
|
Yuzhalin AE. Parallels between the extracellular matrix roles in developmental biology and cancer biology. Semin Cell Dev Biol 2021; 128:90-102. [PMID: 34556419 DOI: 10.1016/j.semcdb.2021.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/28/2022]
Abstract
Interaction of a tumor with its microenvironment is an emerging field of investigation, and the crosstalk between cancer cells and the extracellular matrix is of particular interest, since cancer patients with abundant and stiff extracellular matrices display a poorer prognosis. At the post-juvenile stage, the extracellular matrix plays predominantly a structural role by providing support to cells and tissues; however, during development, matrix proteins exert a plethora of diverse signals to guide the movement and determine the fate of pluripotent cells. Taking a closer look at the communication between the extracellular matrix and cells of a developing body may bring new insights into cancer biology and identify cancer weaknesses. This review discusses parallels between the extracellular matrix roles during development and tumor growth.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Swaminathan G, Kamyabi N, Carter HE, Rajan A, Karandikar U, Criss ZK, Shroyer NF, Robertson MJ, Coarfa C, Huang C, Shannon TE, Tadros M, Estes MK, Maresso AW, Grande-Allen KJ. Effect of substrate stiffness on human intestinal enteroids' infectivity by enteroaggregative Escherichia coli. Acta Biomater 2021; 132:245-259. [PMID: 34280559 PMCID: PMC8434991 DOI: 10.1016/j.actbio.2021.07.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/24/2021] [Accepted: 07/12/2021] [Indexed: 01/08/2023]
Abstract
Human intestinal enteroids (HIE) models have contributed significantly to our understanding of diarrheal diseases and other intestinal infections, but their routine culture conditions fail to mimic the mechanical environment of the native intestinal wall. Because the mechanical characteristics of the intestine significantly alter how pathogens interact with the intestinal epithelium, we used different concentrations of polyethylene glycol (PEG) to generate soft (~2 kPa), medium (~10 kPa), and stiff (~100 kPa) hydrogel biomaterial scaffolds. The height of HIEs cultured in monolayers atop these hydrogels was 18 µm whereas HIEs grown on rigid tissue culture surfaces (with stiffness in the GPa range) were 10 µm. Substrate stiffness also influenced the amount of enteroaggregative E. coli (EAEC strain 042) adhered to the HIEs. We quantified a striking difference in adherence pattern; on the medium and soft gels, the bacteria formed clusters of > 100 and even > 1000 on both duodenal and jejunal HIEs (such as would be found in biofilms), but did not on glass slides and stiff hydrogels. All hydrogel cultured HIEs showed significant enrichment for gene and signaling pathways related to epithelial differentiation, cell junctions and adhesions, extracellular matrix, mucins, and cell signaling compared to the HIEs cultured on rigid tissue culture surfaces. Collectively, these results indicate that the HIE monolayers cultured on the hydrogels are primed for a robust engagement with their mechanical environment, and that the soft hydrogels promote the formation of larger EAEC aggregates, likely through an indirect differential effect on mucus. STATEMENT OF SIGNIFICANCE: Enteroids are a form of in vitro experimental mini-guts created from intestinal stem cells. Enteroids are usually cultured in 3D within Matrigel atop rigid glass or plastic substrates, which fail to mimic the native intestinal mechanical environment. Because intestinal mechanics significantly alter how pathogens interact with the intestinal epithelium, we grew human intestinal enteroids in 2D atop polyethylene glycol (PEG) hydrogel scaffolds that were soft, medium, or stiff. Compared with enteroids grown in 2D atop glass or plastic, the enteroids grown on hydrogels were taller and more enriched in mechanobiology-related gene signaling pathways. Additionally, enteroids on the softest hydrogels supported adhesion of large aggregates of enteroaggregative E. coli. Thus, this platform offers a more biomimetic model for studying enteric diseases.
Collapse
Affiliation(s)
- Ganesh Swaminathan
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States
| | - Nabiollah Kamyabi
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States
| | - Hannah E Carter
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Umesh Karandikar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Zachary K Criss
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States
| | - Noah F Shroyer
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States
| | - Matthew J Robertson
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Cristian Coarfa
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States; Department of Molecular and Cellular Biology - Molecular Regulation, Baylor College of Medicine, Houston, TX, United States
| | - Chenlin Huang
- Department of Biosciences, Rice University, Houston, TX, United States
| | - Tate E Shannon
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States
| | - Madeleine Tadros
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, United States
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Anthony W Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - K Jane Grande-Allen
- Departmcnt of Bioengineering, Rice University, 6100 Main St., MS 142, Houston, TX, United States.
| |
Collapse
|
14
|
Yao X, Wang X, Ding J. Exploration of possible cell chirality using material techniques of surface patterning. Acta Biomater 2021; 126:92-108. [PMID: 33684535 DOI: 10.1016/j.actbio.2021.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Consistent left-right (LR) asymmetry or chirality is critical for embryonic development and function maintenance. While chirality on either molecular or organism level has been well established, that on the cellular level has remained an open question for a long time. Although it remains unclear whether chirality exists universally on the cellular level, valuable efforts have recently been made to explore this fundamental topic pertinent to both cell biology and biomaterial science. The development of material fabrication techniques, surface patterning, in particular, has afforded a unique platform to study cell-material interactions. By using patterning techniques, chirality on the cellular level has been examined for cell clusters and single cells in vitro in well-designed experiments. In this review, we first introduce typical fabrication techniques of surface patterning suitable for cell studies and then summarize the main aspects of preliminary evidence of cell chirality on patterned surfaces to date. We finally indicate the limitations of the studies conducted thus far and describe the perspectives of future research in this challenging field. STATEMENT OF SIGNIFICANCE: While both biomacromolecules and organisms can exhibit chirality, it is not yet conclusive whether a cell has left-right (LR) asymmetry. It is important yet challenging to study and reveal the possible existence of cell chirality. By using the technique of surface patterning, the recent decade has witnessed progress in the exploration of possible cell chirality within cell clusters and single cells. Herein, some important preliminary evidence of cell chirality is collected and analyzed. The open questions and perspectives are also described to promote further investigations of cell chirality in biomaterials.
Collapse
|
15
|
Crespo PV, Campos F, Leal M, Maraver F. Effects of Sodium Chloride-Rich Mineral Water on Intestinal Epithelium. Experimental Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18063261. [PMID: 33809886 PMCID: PMC8004238 DOI: 10.3390/ijerph18063261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/20/2021] [Accepted: 03/19/2021] [Indexed: 12/16/2022]
Abstract
Since knowledge concerning the cellular and tissue substrate that explains the therapeutic action of mineral waters is generally very scarce, we address the different effects that Lanjarón-Capuchina mineral water exerts on the intestinal epithelium in an experimental model as a prototype of the sodium chloride-rich mineral waters used in digestive disorders. In the experimental protocol, two groups of five adult Wistar rats received unrestricted mineral water in their diet or mineral water directly into the gastrointestinal tract through a catheter. A third control group was given a standard diet and water ad libitum. Intestinal samples for scanning electron microscopy were analyzed according to standardized methods. The observations carried out by microscope after the administration of the sodium chloride-rich mineral water clearly indicate that the hypertonic action of this mineral water affects the structure of the intestinal epithelium. It modifies the microvilli absorption in terms of the groups of enterocytes and the secretion of goblet cells, but it particularly affects the epithelial renewal process, accelerating and stimulating cell extrusion. The type of extrusion mechanism observed by microscope allows us to affirm that, although this increased after direct administration, it does not generate an epithelial disruption as it occurs in other circumstances with other extrusion modalities.
Collapse
Affiliation(s)
- Pascual-Vicente Crespo
- Tissue Engineering Group, Department of Histology, University of Granada, 18016 Granada, Spain; (P.-V.C.); (F.C.)
| | - Fernando Campos
- Tissue Engineering Group, Department of Histology, University of Granada, 18016 Granada, Spain; (P.-V.C.); (F.C.)
| | - Manuel Leal
- Professional School of Medical Hydrology, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain;
| | - Francisco Maraver
- Professional School of Medical Hydrology, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain;
- Medical Hydrology Group, Department of Radiology, Rehabilitation & Physiotherapy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
16
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Differentiated Caco-2 cell models in food-intestine interaction study: Current applications and future trends. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2020.11.015] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Chamniansawat S, Kampuang N, Suksridechacin N, Thongon N. Ultrastructural intestinal mucosa change after prolonged inhibition of gastric acid secretion by omeprazole in male rats. Anat Sci Int 2021; 96:142-156. [PMID: 32931001 DOI: 10.1007/s12565-020-00572-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Omeprazole is a potent inhibitor of gastric acid secretion. It was reported that omeprazole induced dramatic gastric mucosa morphologic changes from the resting state to the stimulated state. However, the effect of omeprazole administration on the ultrastructure and absorptive function of small intestines was largely unknown. Here, male Sprague-Dawley rats were daily treated with a single dose of omeprazole for 12 or 24 weeks. Ultrastructure intestinal mucosal change in duodenum, jejunum, and ileum was observed. We also determined small intestine inflammation, using intraepithelial lymphocytes activation. Finally, magnesium levels were measured in plasma, urine, feces, muscle, and bone to determine systemic magnesium balance. Omeprazole-treated rats had significantly decreased the width of tight junction, villous length, and absorptive area of duodenum, jejunum, and ileum compared to control rats. The small intestine of the omeprazole-treated group showed significantly higher intraepithelial lymphocytes activation levels compared with the control group. Lower secretory granules of Paneth cells at the base of the crypts were showed in omeprazole-treated rats. They also had significantly lower plasma, urinary, bone, and muscle Mg2+ contents indicating hypomagnesemia with systemic magnesium deficiency. In conclusion, prolonged omeprazole treatment-induced small intestinal inflammation and villous atrophy, which led to decrease small intestinal magnesium absorption in the condition of proton pump inhibitor-induced hypomagnesemia.
Collapse
Affiliation(s)
- Siriporn Chamniansawat
- Division of Anatomy, Department of Biomedical Sciences, Faculty of Allied Health Sciences, Burapha University, 169 Long-Hard Bangsaen Rd., Saensook, Muang, Chon Buri, 20131, Thailand
| | - Nattida Kampuang
- Division of Physiology, Department of Biomedical Sciences, Faculty of Allied Health Sciences, Burapha University, Chon Buri, Thailand
| | - Nasisorn Suksridechacin
- Division of Physiology, Department of Biomedical Sciences, Faculty of Allied Health Sciences, Burapha University, Chon Buri, Thailand
| | - Narongrit Thongon
- Division of Physiology, Department of Biomedical Sciences, Faculty of Allied Health Sciences, Burapha University, Chon Buri, Thailand.
| |
Collapse
|
19
|
De Gregorio V, La Rocca A, Urciuolo F, Annunziata C, Tornesello ML, Buonaguro FM, Netti PA, Imparato G. Modeling the epithelial-mesenchymal transition process in a 3D organotypic cervical neoplasia. Acta Biomater 2020; 116:209-222. [PMID: 32911106 DOI: 10.1016/j.actbio.2020.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 01/03/2023]
Abstract
Here, we proposed an innovative organotypic cervical tumor model able to investigate the bi-directional crosstalk between epithelium and stroma as well as the key disease features of the epithelial-mesenchymal transition (EMT) process in vitro. By using a modular tissue assembling approach, we developed 3D cervical stromal models composed of primary human cervical fibroblasts (HCFs) or cervical cancer-associated fibroblasts (CCAFs) embedded in their own ECM to produce 3D normal cervical-instructed stroma (NCIS) or 3D cervical cancer-instructed stroma (CCIS), respectively. Then, we demonstrate the role of the tumor microenvironment (TME) in potentiating the intrinsic invasive attitude of cervical cancer derived SiHa cells and increasing their early viral gene expression by comparing the SiHa behavior when cultured on NCIS or CCIS (SiHa-NCIS or SiHa-CCIS). We proved the crucial role of the CCAFs and stromal microenvironment in the mesenchymalization of the cancer epithelial cells by analyzing several EMT markers. We further assessed the expression of the epithelial adhesion molecules, matricellular enzymes, non-collagenous proteins as well as ECM remodeling in terms of collagen fibers texture and assembly. This cervical tumor model, closely recapitulating key cervical carcinogenesis features, may provide efficient and relevant support to current approaches characterizing cancer progression and develop new anticancer therapy targeting stroma rather than cancer cells.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy; Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - Alessia La Rocca
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy; Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, Naples, Italy
| | - Francesco Urciuolo
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy; Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, Naples, Italy
| | - Clorinda Annunziata
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Paolo Antonio Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy; Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy; Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy.
| |
Collapse
|
20
|
Gjorevski N, Avignon B, Gérard R, Cabon L, Roth AB, Bscheider M, Moisan A. Neutrophilic infiltration in organ-on-a-chip model of tissue inflammation. LAB ON A CHIP 2020; 20:3365-3374. [PMID: 32761043 DOI: 10.1039/d0lc00417k] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The multiphasic etiology of tissue inflammation and the fundamental immunological differences between species render inflammatory pathologies difficult to recapitulate in animal models, and account for the paucity of therapies that are successfully translated from rodents to humans. Here, we present a human-relevant organ-on-a-chip platform for experimental inflammatory diseases. We created an immunocompetent in vitro gut model by incorporating intestinal epithelial and immune cells into microfluidic chambers that permit cell movement across an extracellular matrix (ECM) and fluidic channels. This is the first model that integrates a mucosal barrier, a three-dimensional ECM, resident and infiltrating immune cells, and simulates a functional crosstalk that ultimately triggers cellular processes representative of inflammation. Under homeostatic conditions, enterocytes form a tight epithelium and subepithelial macrophages are non-activated. Introduction of pro-inflammatory mediators triggers macrophage activation and inflammation-induced intestinal barrier leakiness. Neutrophils in a parallel, matrix-separated non-epithelial channel are attracted by such a pro-inflammatory microenvironment and migrate through the extracellular matrix, further exacerbating tissue inflammation and damage. With this model, we provide the foundations to recapitulate and investigate the onset of tissue inflammation in a controlled, human-relevant system.
Collapse
Affiliation(s)
- Nikolce Gjorevski
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
21
|
De Gregorio V, Telesco M, Corrado B, Rosiello V, Urciuolo F, Netti PA, Imparato G. Intestine-Liver Axis On-Chip Reveals the Intestinal Protective Role on Hepatic Damage by Emulating Ethanol First-Pass Metabolism. Front Bioeng Biotechnol 2020; 8:163. [PMID: 32258006 PMCID: PMC7090126 DOI: 10.3389/fbioe.2020.00163] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/18/2020] [Indexed: 12/23/2022] Open
Abstract
Intestine-Liver-on-chip systems can be useful to predict oral drug administration and first-pass metabolism in vitro in order to partly replace the animal model. While organ-on-chip technology can count on sophisticated micro-physiological devices, the engineered organs still remain artificial surrogates of the native counterparts. Here, we used a bottom-up tissue engineering strategy to build-up physiologically functional 3D Human Intestine Model (3D-HIM) as well as 3D Liver-microtissues (HepG2-μTPs) in vitro and designed a microfluidic Intestine-Liver-On-Chip (InLiver-OC) to emulate first-pass mechanism occurring in vivo. Our results highlight the ethanol-induced 3D-HIM hyper-permeability and stromal injury, the intestinal prevention on the liver injury, as well as the synergic contribution of the two 3D tissue models on the release of metabolic enzymes after high amount of ethanol administration.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Mariarosaria Telesco
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - Brunella Corrado
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - Valerio Rosiello
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Francesco Urciuolo
- 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
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) University of Naples Federico II, Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| |
Collapse
|
22
|
Kim W, Kim GH. An intestinal model with a finger-like villus structure fabricated using a bioprinting process and collagen/SIS-based cell-laden bioink. Am J Cancer Res 2020; 10:2495-2508. [PMID: 32194815 PMCID: PMC7052892 DOI: 10.7150/thno.41225] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023] Open
Abstract
The surface of the small intestine has a finger-like microscale villus structure, which provides a large surface area to realize efficient digestion and absorption. However, the fabrication of a villus structure using a cell-laden bioink containing a decellularized small intestine submucosa, SIS, which can induce significant cellular activities, has not been attempted owing to the limited mechanical stiffness, which sustains the complex projective finger-like 3D structure. In this work, we developed a human intestinal villi model with an innovative bioprinting process using a collagen/SIS cell-laden bioink. Methods: A Caco-2-laden microscale villus structure (geometry of the villus: height = 831.1 ± 36.2 μm and diameter = 190.9 ± 3.9 μm) using a bioink consisting of collagen type-I and SIS was generated using a vertically moving 3D bioprinting process. By manipulating various compositions of dECM and a crosslinking agent in the bioink and the processing factors (printing speed, printing time, and pneumatic pressure), the villus structure was achieved. Results: The epithelial cell-laden collagen/SIS villi showed significant cell proliferation (1.2-fold) and demonstrated meaningful results for the various cellular activities, such as the expression of tight-junction proteins (ZO-1 and E-cadherin), ALP and ANPEP activities, MUC17 expression, and the permeability coefficient and the glucose uptake ability, compared with the pure 3D collagen villus structure. Conclusion: In vitro cellular activities demonstrated that the proposed cell-laden collagen/dECM villus structure generates a more meaningful epithelium layer mimicking the intestinal structure, compared with the pure cell-laden collagen villus structure having a similar villus geometry. Based on the results, we believe that this dECM-based 3D villus model will be helpful in obtaining a more realistic physiological small-intestine model.
Collapse
|
23
|
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
|
24
|
Sibilio S, De Gregorio V, Urciuolo F, Netti P, Imparato G. Effect of peristaltic-like movement on bioengineered intestinal tube. Mater Today Bio 2019; 4:100027. [PMID: 32159155 PMCID: PMC7061615 DOI: 10.1016/j.mtbio.2019.100027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/05/2019] [Accepted: 09/01/2019] [Indexed: 01/06/2023] Open
Abstract
The intestine is a highly heterogeneous hollow organ with biological, mechanical and chemical differences between lumen and wall. A functional human intestine model able to recreate the in vivo dynamic nature as well as the native tissue morphology is demanded for disease research and drug discovery. Here, we present a system, which combines an engineered three-dimensional (3D) tubular-shaped intestine model (3D In-tube) with a custom-made microbioreactor to impart the key aspects of the in vivo microenvironment of the human intestine, mimicking the rhythmic peristaltic movement. We adapted a previously established bottom-up tissue engineering approach, to produce the 3D tubular-shaped lamina propria and designed a glass microbioreactor to induce the air-liquid interface condition and peristaltic-like motion. Our results demonstrate the production of a villi-like protrusion and a correct spatial differentiation of the intestinal epithelial cells in enterocyte-like as well as mucus-producing-like cells on the lumen side of the 3D In-tube. This dynamic platform offers a proof-of-concept model of the human intestine.
Collapse
Affiliation(s)
- S. Sibilio
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, P.le Tecchio 80, Naples, 80125, Italy
| | - V. De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, Naples, 80125, Italy
| | - F. Urciuolo
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, P.le Tecchio 80, Naples, 80125, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, Naples, 80125, Italy
| | - P.A. Netti
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, P.le Tecchio 80, Naples, 80125, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, Naples, 80125, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, Naples, 80125, Italy
| | - G. Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, Naples, 80125, Italy
| |
Collapse
|
25
|
Charlesworth RPG, Marsh MN. From 2-dimensional to 3-dimensional: Overcoming dilemmas in intestinal mucosal interpretation. World J Gastroenterol 2019; 25:2402-2415. [PMID: 31171885 PMCID: PMC6543240 DOI: 10.3748/wjg.v25.i20.2402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/04/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
The purpose of this review is to provide a definitive account of small intestinal mucosal structure and interpretation. The coeliac lesion has been well known, but not well described to date and this review aims to identify the interpretative difficulties which have arisen over time with the histological assessment of coeliac disease. In early coeliac interpretation, there were significant inaccuracies, particularly surrounding intraepithelial lymphocyte counts and the degree of villous flattening which occurred in the tissue. Many of these interpretive pitfalls are still encountered today, increasing the potential for diagnostic errors. These difficulties are mostly due to the fact that stained 2-dimensional sections can never truly represent the 3-dimensional framework of the intestinal tissue under investigation. Therefore, this review offers a critical account occasioned by these 2-dimensional interpretative errors and which, in our opinion, should in general be jettisoned. As a result, we leave a framework regarding the true 3-dimensional knowledge of mucosal structure accrued over the 70-year period of study, and one which is available for future reference.
Collapse
Affiliation(s)
| | - Michael N Marsh
- Wolfson College, University of Oxford, Oxford OX2 6UD, United Kingdom
| |
Collapse
|
26
|
A three‐dimensional microfluidized liver system to assess hepatic drug metabolism and hepatotoxicity. Biotechnol Bioeng 2019; 116:1152-1163. [DOI: 10.1002/bit.26902] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022]
|