1
|
Wu H, Mu C, Xu L, Yu K, Shen L, Zhu W. Host-microbiota interaction in intestinal stem cell homeostasis. Gut Microbes 2024; 16:2353399. [PMID: 38757687 PMCID: PMC11110705 DOI: 10.1080/19490976.2024.2353399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
Intestinal stem cells (ISCs) play a pivotal role in gut physiology by governing intestinal epithelium renewal through the precise regulation of proliferation and differentiation. The gut microbiota interacts closely with the epithelium through myriad of actions, including immune and metabolic interactions, which translate into tight connections between microbial activity and ISC function. Given the diverse functions of the gut microbiota in affecting the metabolism of macronutrients and micronutrients, dietary nutrients exert pronounced effects on host-microbiota interactions and, consequently, the ISC fate. Therefore, understanding the intricate host-microbiota interaction in regulating ISC homeostasis is imperative for improving gut health. Here, we review recent advances in understanding host-microbiota immune and metabolic interactions that shape ISC function, such as the role of pattern-recognition receptors and microbial metabolites, including lactate and indole metabolites. Additionally, the diverse regulatory effects of the microbiota on dietary nutrients, including proteins, carbohydrates, vitamins, and minerals (e.g. iron and zinc), are thoroughly explored in relation to their impact on ISCs. Thus, we highlight the multifaceted mechanisms governing host-microbiota interactions in ISC homeostasis. Insights gained from this review provide strategies for the development of dietary or microbiota-based interventions to foster gut health.
Collapse
Affiliation(s)
- Haiqin Wu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Chunlong Mu
- Food Informatics, AgResearch, Te Ohu Rangahau Kai, Palmerston North, New Zealand
| | - Laipeng Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Le Shen
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
2
|
Yin Y, Xu L, Steinway SN. Editorial: 3D organoid and organ-on-a-chip and their applications for virology and antiviral research. Front Microbiol 2023; 14:1266136. [PMID: 37614596 PMCID: PMC10443217 DOI: 10.3389/fmicb.2023.1266136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Affiliation(s)
- Yuebang Yin
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, CE Rotterdam, Rotterdam, Netherlands
| | - Lei Xu
- National Key Laboratory of Crop Improvement for Stress Tolerance and Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Xianyang, Shaanxi, China
| | | |
Collapse
|
3
|
Li Y, Li P, Yu X, Zheng X, Gu Q. Exploitation of In Vivo-Emulated In Vitro System in Advanced Food Science. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37023249 DOI: 10.1021/acs.jafc.2c07289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Reasonable model construction contributes to the accuracy of experimental results. Multiple in vivo models offer reliable choices for effective evaluation, whereas their applications are hampered due to adverse features including high time-consumption, high cost and ethical contradictions. In vivo-emulated in vitro systems (IVE systems) have experienced rapid development and have been brought into food science for about two decades. IVE systems' flexibly gathers the strengths of in vitro and in vivo models into one, reflecting the results in an efficient, systematic and interacted manner. In this review, we comprehensively reviewed the current research progress of IVE systems based on the literature published in the recent two decades. By categorizing the IVE systems into 2D coculture models, spheroids and organoids, their applications were systematically summarized and typically exemplified. The pros and cons of IVE systems were also thoroughly discussed, drawing attention to present challenges and inspiring potential orientation and future perspectives. The wide applicability and multiple possibilities suggest IVE systems as an effective and persuasive platform in the future of advanced food science.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, 310018, Zhejiang, People's Republic of China
| | - Ping Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, 310018, Zhejiang, People's Republic of China
| | - Xin Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, People's Republic of China
- Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, and National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, People's Republic of China
- Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, and National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, 310018, Zhejiang, People's Republic of China
| |
Collapse
|
4
|
Butts M, Sundaram VL, Murughiyan U, Borthakur A, Singh S. The Influence of Alcohol Consumption on Intestinal Nutrient Absorption: A Comprehensive Review. Nutrients 2023; 15:nu15071571. [PMID: 37049411 PMCID: PMC10096942 DOI: 10.3390/nu15071571] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Chronic alcohol use has been attributed to the development of malnutrition. This is in part due to the inhibitory effect of ethanol on the absorption of vital nutrients, including glucose, amino acids, lipids, water, vitamins, and minerals within the small intestine. Recent advances in research, along with new cutting-edge technologies, have advanced our understanding of the mechanism of ethanol's effect on intestinal nutrient absorption at the brush border membrane (BBM) of the small intestine. However, further studies are needed to delineate how ethanol consumption could have an impact on altered nutrient absorption under various disease conditions. Current research has elucidated the relationship of alcohol consumption on glucose, glutamine, vitamins B1 (thiamine), B2 (riboflavin), B9 (folate), C (ascorbic acid), selenium, iron, and zinc absorption within the small intestine. We conducted systematic computerized searches in PubMed using the following keywords: (1) "Alcohol effects on nutrient transport"; (2) "Alcohol mediated malabsorption of nutrients"; (3) "Alcohol effects on small intestinal nutrient transport"; and (4) "Alcohol mediated malabsorption of nutrients in small intestine". We included the relevant studies in this review. The main objective of this review is to marshal and analyze previously published research articles and discuss, in-depth, the understanding of ethanol's effect in modulating absorption of vital macro and micronutrients in health and disease conditions. This could ultimately provide great insights in the development of new therapeutic strategies to combat malnutrition associated with alcohol consumption.
Collapse
Affiliation(s)
- Molly Butts
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Vijaya Lakshmi Sundaram
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Usha Murughiyan
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Alip Borthakur
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Soudamani Singh
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| |
Collapse
|
5
|
Nie J, Liao W, Zhang Z, Zhang M, Wen Y, Capanoglu E, Sarker MMR, Zhu R, Zhao C. A 3D co-culture intestinal organoid system for exploring glucose metabolism. Curr Res Food Sci 2022; 6:100402. [DOI: 10.1016/j.crfs.2022.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/02/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
|
6
|
Günther C, Winner B, Neurath MF, Stappenbeck TS. Organoids in gastrointestinal diseases: from experimental models to clinical translation. Gut 2022; 71:1892-1908. [PMID: 35636923 PMCID: PMC9380493 DOI: 10.1136/gutjnl-2021-326560] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
We are entering an era of medicine where increasingly sophisticated data will be obtained from patients to determine proper diagnosis, predict outcomes and direct therapies. We predict that the most valuable data will be produced by systems that are highly dynamic in both time and space. Three-dimensional (3D) organoids are poised to be such a highly valuable system for a variety of gastrointestinal (GI) diseases. In the lab, organoids have emerged as powerful systems to model molecular and cellular processes orchestrating natural and pathophysiological human tissue formation in remarkable detail. Preclinical studies have impressively demonstrated that these organs-in-a-dish can be used to model immunological, neoplastic, metabolic or infectious GI disorders by taking advantage of patient-derived material. Technological breakthroughs now allow to study cellular communication and molecular mechanisms of interorgan cross-talk in health and disease including communication along for example, the gut-brain axis or gut-liver axis. Despite considerable success in culturing classical 3D organoids from various parts of the GI tract, some challenges remain to develop these systems to best help patients. Novel platforms such as organ-on-a-chip, engineered biomimetic systems including engineered organoids, micromanufacturing, bioprinting and enhanced rigour and reproducibility will open improved avenues for tissue engineering, as well as regenerative and personalised medicine. This review will highlight some of the established methods and also some exciting novel perspectives on organoids in the fields of gastroenterology. At present, this field is poised to move forward and impact many currently intractable GI diseases in the form of novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Stem Cell Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Yin L, Li J, Zhang Y, Yang Q, Yang C, Yi Z, Yin Y, Wang Q, Li J, Ding N, Zhang Z, Yang H, Yin Y. Changes in progenitors and differentiated epithelial cells of neonatal piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:265-276. [PMID: 34988308 PMCID: PMC8693152 DOI: 10.1016/j.aninu.2021.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 01/13/2023]
Abstract
This study aimed to assess the changes of small intestinal morphology, progenitors, differentiated epithelial cells, and potential mechanisms in neonatal piglets. Hematoxylin and eosin staining of samples from 36 piglets suggested that dramatic changes were observed in the jejunum crypts depth and crypt fission index of neonatal piglets (P < 0.001). The number of intestinal stem cells (ISC) tended to increase (P < 0.10), and a decreased number of enteroendocrine cells appeared in the jejunal crypt on d 7 (P < 0.05). Furthermore, the mRNA expression of jejunal chromogranin A (ChgA) was down-regulated in d 7 piglets (P < 0.05). There was an up-regulation of the adult ISC marker gene of SPARC related modular calcium binding 2 (Smoc2), and Wnt/β-catenin target genes on d 7 (P < 0.05). These results were further verified in vitro enteroid culture experiments. A mass of hollow spheroids was cultured from the fetal intestine of 0-d-old piglets (P < 0.001), whereas substantial organoids with budding and branching structures were cultured from the intestine of 7-d-old piglets (P < 0.001). The difference was reflected by the organoid budding efficiency, crypt domains per organoid, and the surface area of the organoid. Furthermore, spheroids on d 0 had more Ki67-positive cells and enteroendocrine cells (P < 0.05) and showed a decreasing trend in the ISC and goblet cells (P < 0.10). Moreover, the mRNA expression of spheroids differed markedly from that of organoids, with low expression of intestinal differentiation gene (Lysozyme; P < 0.05), epithelial-specific markers (Villin, E-cadherin; P < 0.05), and adult ISC markers (leucine-rich repeat-containing G protein-coupled receptor 5 [Lgr5], Smoc2; P < 0.001), and up-regulation of fetal marker (connexin 43 [Cnx43]; P < 0.05). The mRNA expression of relevant genes was up-regulated, and involved in Wnt/β-catenin, epidermal growth factor (EGF), Notch, and bone morphogenetic protein (BMP) signaling on d 7 organoids (P < 0.05). Spheroids displayed low differentiated phenotype and high proliferation, while organoids exhibited strong differentiation potential. These results indicated that the conversion from the fetal progenitors (spheroids) to adult ISC (normal organoids) might largely be responsible for the fast development of intestinal epithelial cells in neonatal piglets.
Collapse
Affiliation(s)
- Lanmei Yin
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China.,National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Jun Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.,State Key Laboratory of Food Safety Technology for Meat Products, Yinxiang Group, Fujian Aonong BiologicaI Science and Technology Group Co., Ltd., Key Laboratory of Swine Nutrition and Feed Science of Fujian Province, Aonong Group, Zhangzhou, Fujian, 363000, China
| | - Yitong Zhang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Qing Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Cuiyan Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Zhenfeng Yi
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Yuebang Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Qiye Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Jianzhong Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China
| | - Nengshui Ding
- State Key Laboratory of Food Safety Technology for Meat Products, Yinxiang Group, Fujian Aonong BiologicaI Science and Technology Group Co., Ltd., Key Laboratory of Swine Nutrition and Feed Science of Fujian Province, Aonong Group, Zhangzhou, Fujian, 363000, China
| | - Zhigang Zhang
- State Key Laboratory of Food Safety Technology for Meat Products, Yinxiang Group, Fujian Aonong BiologicaI Science and Technology Group Co., Ltd., Key Laboratory of Swine Nutrition and Feed Science of Fujian Province, Aonong Group, Zhangzhou, Fujian, 363000, China
| | - Huansheng Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Yulong Yin
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China.,Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China.,National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| |
Collapse
|
8
|
Co JY, Margalef-Català M, Monack DM, Amieva MR. Controlling the polarity of human gastrointestinal organoids to investigate epithelial biology and infectious diseases. Nat Protoc 2021; 16:5171-5192. [PMID: 34663962 PMCID: PMC8841224 DOI: 10.1038/s41596-021-00607-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/19/2021] [Indexed: 02/08/2023]
Abstract
Human epithelial organoids-3D spheroids derived from adult tissue stem cells-enable investigation of epithelial physiology and disease and host interactions with microorganisms, viruses and bioactive molecules. One challenge in using organoids is the difficulty in accessing the apical, or luminal, surface of the epithelium, which is enclosed within the organoid interior. This protocol describes a method we previously developed to control human and mouse organoid polarity in suspension culture such that the apical surface faces outward to the medium (apical-out organoids). Our protocol establishes apical-out polarity rapidly (24-48 h), preserves epithelial integrity, maintains secretory and absorptive functions and allows regulation of differentiation. Here, we provide a detailed description of the organoid polarity reversal method, compatible characterization assays and an example of an application of the technology-specifically the impact of host-microbe interactions on epithelial function. Control of organoid polarity expands the possibilities of organoid use in gastrointestinal and respiratory health and disease research.
Collapse
Affiliation(s)
- Julia Y Co
- Division of Infectious Diseases, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Mar Margalef-Català
- Division of Infectious Diseases, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Denise M Monack
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Manuel R Amieva
- Division of Infectious Diseases, Department of Pediatrics, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
9
|
Yin L, Yang Q, Zhang Y, Wan D, Yin Y, Wang Q, Huang J, Li J, Yang H, Yin Y. Dietary Copper Improves Intestinal Morphology via Modulating Intestinal Stem Cell Activity in Pigs. Animals (Basel) 2021; 11:2513. [PMID: 34573479 PMCID: PMC8471658 DOI: 10.3390/ani11092513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022] Open
Abstract
Copper (Cu) is an essential micronutrient for animals. Many studies have been conducted on the effects of dietary Cu on growth performance, intestinal morphology, and function of piglets. However, the underlying mechanism remains to be explored. Intestinal stem cells (ISC) drive the development and constant renewal of intestinal epithelium. Therefore, we hypothesized that dietary Cu affects piglets' intestinal development via modulating ISC activity. A total of eighty-five 21-day-old piglets were randomly assigned to five groups, where 25, 50, 75, 100, and 125 mg CuSO4/kg on a dry matter basis were supplemented to the basal diet at phase 1 (day 0 to 21). Increasing the dietary Cu concentration decreased (p < 0.05) villus width but increased (p < 0.001) the number of Ki67-positive cells. At phase 2 (day 22 to 163), the other 45 pigs were offered the same diets. Villus height in the 125 mg/kg Cu group was greater (p < 0.001) than in the other groups. Moreover, the effects of Cu on ISC activity in vitro were tested to explore the underlying mechanism. Compared to the control group, 10 μmol/L CuSO4·5H2O increased (p < 0.001) the organoid budding efficiency, crypt depth, and crypts per organoid. Dietary Cu improved the intestinal morphology of finishing pigs via promoting cell proliferation and modulating ISC activity.
Collapse
Affiliation(s)
- Lanmei Yin
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
| | - Qing Yang
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
| | - Yiming Zhang
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (D.W.); (Y.Y.)
| | - Dan Wan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (D.W.); (Y.Y.)
| | - Yuebang Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (D.W.); (Y.Y.)
| | - Qiye Wang
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
| | - Jing Huang
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
| | - Jianzhong Li
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Huansheng Yang
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yulong Yin
- Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (L.Y.); (Q.Y.); (Y.Z.); (Q.W.); (J.H.); (J.L.); (Y.Y.)
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (D.W.); (Y.Y.)
| |
Collapse
|
10
|
Zhang Y, Huang S, Zhong W, Chen W, Yao B, Wang X. 3D organoids derived from the small intestine: An emerging tool for drug transport research. Acta Pharm Sin B 2021; 11:1697-1707. [PMID: 34386316 PMCID: PMC8343122 DOI: 10.1016/j.apsb.2020.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Small intestine in vitro models play a crucial role in drug transport research. Although conventional 2D cell culture models, such as Caco-2 monolayer, possess many advantages, they should be interpreted with caution because they have relatively poor physiologically reproducible phenotypes and functions. With the development of 3D culture technology, pluripotent stem cells (PSCs) and adult somatic stem cells (ASCs) show remarkable self-organization characteristics, which leads to the development of intestinal organoids. Based on previous studies, this paper reviews the application of intestinal 3D organoids in drug transport mediated by P-glycoprotein (P-gp), breast cancer resistance protein (BCRP) and multidrug resistance protein 2 (MRP2). The advantages and limitations of this model are also discussed. Although there are still many challenges, intestinal 3D organoid model has the potential to be an excellent tool for drug transport research.
Collapse
Key Words
- 3D organoid
- ASCs, adult somatic stem cells
- BCRP, breast cancer resistance protein
- BMP, bone morphogenetic protein
- CDF, 5(6)-carboxy-2′,7′-dichlorofluorescein
- Caco-2 cell monolayer
- DDI, drug–drug interactions
- Drug transporter
- EGF, epidermal growth factor
- ER, efflux ratio
- ESCs, embryonic stem cells
- FGF, fibroblast growth factor
- Lgr5+, leucine-rich-repeat-containing G-protein-coupled receptor 5 positive
- MCT, monocarboxylate transporter protein
- MRP2, multidrug resistance protein 2
- NBD, nucleotide-binding domain
- OATP, organic anion transporting polypeptide
- OCT, organic cation transporter
- OCTN, carnitine/organic cation transporter
- P-glycoprotein
- P-gp, P-glycoprotein
- PEPT, peptide transporter protein
- PMAT, plasma membrane monoamine transporter
- PSCs, pluripotent stem cells
- Papp, apparent permeability coefficient
- Rh123, rhodamine 123
- SLC, solute carrier
- Small intestine
- TEER, transepithelial electrical resistance
- TMDs, transmembrane domains
- cMOAT, canalicular multispecific organic anion transporter
- iPSCs, induced pluripotent stem cells
Collapse
Affiliation(s)
- Yuanjin Zhang
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Weiguo Zhong
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
| | - Wenxia Chen
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
- Corresponding author. Tel.: +86 21 2420 6564; fax: +86 21 5434 4922.
| |
Collapse
|
11
|
Shurson GC, Hung YT, Jang JC, Urriola PE. Measures Matter-Determining the True Nutri-Physiological Value of Feed Ingredients for Swine. Animals (Basel) 2021; 11:1259. [PMID: 33925594 PMCID: PMC8146707 DOI: 10.3390/ani11051259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 01/10/2023] Open
Abstract
Many types of feed ingredients are used to provide energy and nutrients to meet the nutritional requirements of swine. However, the analytical methods and measures used to determine the true nutritional and physiological ("nutri-physiological") value of feed ingredients affect the accuracy of predicting and achieving desired animal responses. Some chemical characteristics of feed ingredients are detrimental to pig health and performance, while functional components in other ingredients provide beneficial health effects beyond their nutritional value when included in complete swine diets. Traditional analytical procedures and measures are useful for determining energy and nutrient digestibility of feed ingredients, but do not adequately assess their true physiological or biological value. Prediction equations, along with ex vivo and in vitro methods, provide some benefits for assessing the nutri-physiological value of feed ingredients compared with in vivo determinations, but they also have some limitations. Determining the digestion kinetics of the different chemical components of feed ingredients, understanding how circadian rhythms affect feeding behavior and the gastrointestinal microbiome of pigs, and accounting for the functional properties of many feed ingredients in diet formulation are the emerging innovations that will facilitate improvements in precision swine nutrition and environmental sustainability in global pork-production systems.
Collapse
Affiliation(s)
- Gerald C. Shurson
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA; (Y.-T.H.); (J.C.J.); (P.E.U.)
| | | | | | | |
Collapse
|
12
|
Gut health: The results of microbial and mucosal immune interactions in pigs. ACTA ACUST UNITED AC 2021; 7:282-294. [PMID: 34258416 PMCID: PMC8245825 DOI: 10.1016/j.aninu.2021.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/09/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
There are a large number of microorganisms in the porcine intestinal tract. These microorganisms and their metabolites contribute to intestinal mucosal immunity, which is of great importance to the health of the host. The host immune system can regulate the distribution and composition of intestinal microorganisms and regulate the homeostasis of intestinal flora by secreting a variety of immune effector factors, such as mucin, secretory immunoglobulin A (sIgA), regenerating islet-derived III (RegIII)γ, and defensin. Conversely, intestinal microorganisms can also promote the differentiation of immune cells including regulatory T cells (Treg) and Th17 cells through their specific components or metabolites. Studies have shown that imbalances in the intestinal flora can lead to bacterial translocation and compromised intestinal barrier function, affecting the health of the body. This review focuses on the composition of the pig intestinal flora and the characteristics of intestinal mucosal immunity, discusses the interaction mechanism between the flora and intestinal mucosal immunity, as well as the regulation through fecal microbiota transplantation (FMT), dietary nutritional composition, probiotics and prebiotics of pig intestinal microecology. Finally, this review provides insights into the relationship between intestinal microorganisms and the mucosal immune system.
Collapse
|
13
|
Shao BZ, Yao Y, Zhai JS, Zhu JH, Li JP, Wu K. The Role of Autophagy in Inflammatory Bowel Disease. Front Physiol 2021; 12:621132. [PMID: 33633585 PMCID: PMC7902040 DOI: 10.3389/fphys.2021.621132] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an idiopathic intestinal inflammatory disease, including ulcerative colitis (UC) and Crohn’s disease (CD). The abnormality of inflammatory and immune responses in the intestine contributes to the pathogenesis and progression of IBD. Autophagy is a vital catabolic process in cells. Recent studies report that autophagy is highly involved in various kinds of diseases, especially inflammation-related diseases, such as IBD. In this review, the biological characteristics of autophagy and its role in IBD will be described and discussed based on recent literature. In addition, several therapies for IBD through modulating the inflammasome and intestinal microbiota taking advantage of autophagy regulation will be introduced. We aim to bring new insight in the exploration of mechanisms for IBD and development of novel therapeutic strategies against IBD.
Collapse
Affiliation(s)
- Bo-Zong Shao
- The 8th Medical Center of General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Yi Yao
- The 8th Medical Center of General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Jun-Shan Zhai
- The 8th Medical Center of General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Jian-Hua Zhu
- The 8th Medical Center of General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Jin-Ping Li
- The 8th Medical Center of General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Kai Wu
- The 8th Medical Center of General Hospital of the Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
14
|
Hao X, Li W. Chloroquine diphosphate suppresses liver cancer via inducing apoptosis in Wistar rats using interventional therapy. Oncol Lett 2021; 21:233. [PMID: 33613722 DOI: 10.3892/ol.2021.12494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/08/2020] [Indexed: 11/05/2022] Open
Abstract
Liver cancer ranks as the second leading cause of cancer-associated mortality worldwide. To date, neither current ablation therapy nor chemotherapy are considered ideal in improving the outcome of liver cancer. Therefore, more effective therapies for treating this devastating disease are urgently required. Interventional therapy has been used for numerous years in the treatment of different types of cancer, and is characterized by the direct delivery of anticancer drugs into the tumor. It has been reported that antimalarial chloroquine diphosphate (CQ) exerts effective anticancer activity against several types of cancer. However, its effect on liver cancer remains unclear. Therefore, in the present study, 2D monolayer cell culture and 3D spheroid in vitro models, and a rat model, were utilized to investigate the effect of CQ on liver cancer. CQ demonstrated an effective anticancer effect on HepG2 cells and 3D liver spheroids. Furthermore, the drug significantly inhibited cell growth and viability in the 2D and 3D in vitro models. The CQ-based intervention treatment effectively attenuated tumor size and weight, increased food intake and consumption of drinking water, and improved body weight and survival rate of rats in the in vivo model. In addition, treatment with CQ potently increased the expression levels of the apoptosis-related genes. Taken together, the findings of the present study may provide a novel insight into the development of safe and effective treatments for liver cancer.
Collapse
Affiliation(s)
- Xiaoguang Hao
- Department of Radiology, The Fourth Hospital of Hebei Medicine University, Shijiazhuang, Hebei 050000, P.R. China
| | - Weijing Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medicine University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
15
|
Yin Y, Liu PY, Shi Y, Li P. Single-Cell Sequencing and Organoids: A Powerful Combination for Modelling Organ Development and Diseases. Rev Physiol Biochem Pharmacol 2021; 179:189-210. [PMID: 33619630 DOI: 10.1007/112_2020_47] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development and function of a particular organ and the pathogenesis of various diseases remain intimately linked to the features of each cell type in the organ. Conventional messenger RNA- or protein-based methodologies often fail to elucidate the contribution of rare cell types, including some subpopulations of stem cells, short-lived progenitors and circulating tumour cells, thus hampering their applications in studies regarding organ development and diseases. The scRNA-seq technique represents a new approach for determining gene expression variability at the single-cell level. Organoids are new preclinical models that recapitulate complete or partial features of their original organ and are thought to be superior to cell models in mimicking the sophisticated spatiotemporal processes of the development and regeneration and diseases. In this review, we highlight recent advances in the field of scRNA-seq, organoids and their current applications and summarize the advantages of using a combination of scRNA-seq and organoid technology to model diseases and organ development.
Collapse
Affiliation(s)
- Yuebang Yin
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Peng-Yu Liu
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yinghua Shi
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.
| | - Ping Li
- State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Tianhe District, Guangzhou, China.
| |
Collapse
|
16
|
Modasia A, Parker A, Jones E, Stentz R, Brion A, Goldson A, Defernez M, Wileman T, Ashley Blackshaw L, Carding SR. Regulation of Enteroendocrine Cell Networks by the Major Human Gut Symbiont Bacteroides thetaiotaomicron. Front Microbiol 2020; 11:575595. [PMID: 33240233 PMCID: PMC7677362 DOI: 10.3389/fmicb.2020.575595] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Gut microbes have critical roles in maintaining host physiology, but their effects on epithelial chemosensory enteroendocrine cells (EEC) remain unclear. We investigated the role that the ubiquitous commensal gut bacterium Bacteriodes thetaiotaomicron (Bt) and its major fermentation products, acetate, propionate, and succinate (APS) have in shaping EEC networks in the murine gastrointestinal tract (GIT). The distribution and numbers of EEC populations were assessed in tissues along the GIT by fluorescent immunohistochemistry in specific pathogen free (SPF), germfree (GF) mice, GF mice conventionalized by Bt or Lactobacillus reuteri (Lr), and GF mice administered APS. In parallel, we also assessed the suitability of using intestinal crypt-derived epithelial monolayer cultures for these studies. GF mice up-regulated their EEC network, in terms of a general EEC marker chromogranin A (ChrA) expression, numbers of serotonin-producing enterochromaffin cells, and both hormone-producing K- and L-cells, with a corresponding increase in serum glucagon-like peptide-1 (GLP-1) levels. Bt conventionalization restored EEC numbers to levels in SPF mice with regional specificity; the effects on ChrA and L-cells were mainly in the small intestine, the effects on K-cells and EC cells were most apparent in the colon. By contrast, Lr did not restore EEC networks in conventionalized GF mice. Analysis of secretory epithelial cell monolayer cultures from whole small intestine showed that intestinal monolayers are variable and with the possible exclusion of GIP expressing cells, did not accurately reflect the EEC cell makeup seen in vivo. Regarding the mechanism of action of Bt on EECs, colonization of GF mice with Bt led to the production and accumulation of acetate, propionate and succinate (APS) in the caecum and colon, which when administered at physiological concentrations to GF mice via their drinking water for 10 days mimicked to a large extent the effects of Bt in GF mice. After withdrawal of APS, the changes in some EEC were maintained and, in some cases, were greater than during APS treatment. This data provides evidence of microbiota influences on regulating EEC networks in different regions of the GIT, with a single microbe, Bt, recapitulating its role in a process that may be dependent upon its fermentation products.
Collapse
Affiliation(s)
- Amisha Modasia
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Aimee Parker
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Emily Jones
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Regis Stentz
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Arlaine Brion
- Core Science Resources, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Andrew Goldson
- Core Science Resources, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Marianne Defernez
- Core Science Resources, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Tom Wileman
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - L. Ashley Blackshaw
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Simon R. Carding
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
17
|
Kong X, Wang X, Qin Y, Han J. Effects of sunset yellow on proliferation and differentiation of intestinal epithelial cells in murine intestinal organoids. J Appl Toxicol 2020; 41:953-963. [DOI: 10.1002/jat.4080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 08/14/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Xiunan Kong
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| | - Xiu Wang
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| | - Yumei Qin
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| | - Jianzhong Han
- School of Food Science and Biotechnology Zhejiang Gongshang University Hangzhou China
| |
Collapse
|
18
|
Cassotta M, Forbes-Hernández TY, Calderón Iglesias R, Ruiz R, Elexpuru Zabaleta M, Giampieri F, Battino M. Links between Nutrition, Infectious Diseases, and Microbiota: Emerging Technologies and Opportunities for Human-Focused Research. Nutrients 2020; 12:E1827. [PMID: 32575399 PMCID: PMC7353391 DOI: 10.3390/nu12061827] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
The interaction between nutrition and human infectious diseases has always been recognized. With the emergence of molecular tools and post-genomics, high-resolution sequencing technologies, the gut microbiota has been emerging as a key moderator in the complex interplay between nutrients, human body, and infections. Much of the host-microbial and nutrition research is currently based on animals or simplistic in vitro models. Although traditional in vivo and in vitro models have helped to develop mechanistic hypotheses and assess the causality of the host-microbiota interactions, they often fail to faithfully recapitulate the complexity of the human nutrient-microbiome axis in gastrointestinal homeostasis and infections. Over the last decade, remarkable progress in tissue engineering, stem cell biology, microfluidics, sequencing technologies, and computing power has taken place, which has produced a new generation of human-focused, relevant, and predictive tools. These tools, which include patient-derived organoids, organs-on-a-chip, computational analyses, and models, together with multi-omics readouts, represent novel and exciting equipment to advance the research into microbiota, infectious diseases, and nutrition from a human-biology-based perspective. After considering some limitations of the conventional in vivo and in vitro approaches, in this review, we present the main novel available and emerging tools that are suitable for designing human-oriented research.
Collapse
Affiliation(s)
- Manuela Cassotta
- Centre for Nutrition and Health, Universidad Europea del Atlántico (UEA), 39001 Santander, Spain; (M.C.); (R.C.I.); (R.R.)
| | - Tamara Yuliett Forbes-Hernández
- Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
| | - Ruben Calderón Iglesias
- Centre for Nutrition and Health, Universidad Europea del Atlántico (UEA), 39001 Santander, Spain; (M.C.); (R.C.I.); (R.R.)
| | - Roberto Ruiz
- Centre for Nutrition and Health, Universidad Europea del Atlántico (UEA), 39001 Santander, Spain; (M.C.); (R.C.I.); (R.R.)
| | - Maria Elexpuru Zabaleta
- Dipartimento di Scienze Cliniche e Molecolari, Facoltà di Medicina, Università Politecnica delle Marche, 60131 Ancona, Italy;
| | - Francesca Giampieri
- Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy
- College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Maurizio Battino
- Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
19
|
Liu YL, Guo SG, Xie CY, Niu K, De Jonge H, Wu X. Uridine inhibits the stemness of intestinal stem cells in 3D intestinal organoids and mice. RSC Adv 2020; 10:6377-6387. [PMID: 35496025 PMCID: PMC9049648 DOI: 10.1039/c9ra07742a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
The activity of intestinal stem cells (ISCs) is foremost in maintaining homeostasis and repair of intestines.
Collapse
Affiliation(s)
- Yi-Lin Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
- Chinese Academy of Sciences
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process
| | - Song-Ge Guo
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
- Chinese Academy of Sciences
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process
| | - Chun-yan Xie
- College of Resources and Environment
- College of Bioscience and Biotechnology
- Hunan Agricultural University
- Changsha
- China
| | - Kaimin Niu
- Institute of Biological Resources
- Jiangxi Academy of Sciences
- Nanchang 330096
- China
| | - Hugo De Jonge
- Department of Gastroenterology and Hepatology
- Erasmus MC University Medical Center
- Rotterdam
- Netherlands
| | - Xin Wu
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
- Chinese Academy of Sciences
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process
| |
Collapse
|
20
|
Dicarlo M, Teti G, Verna G, Liso M, Cavalcanti E, Sila A, Raveenthiraraj S, Mastronardi M, Santino A, Serino G, Lippolis A, Sobolewski A, Falconi M, Chieppa M. Quercetin Exposure Suppresses the Inflammatory Pathway in Intestinal Organoids from Winnie Mice. Int J Mol Sci 2019; 20:ijms20225771. [PMID: 31744123 PMCID: PMC6888448 DOI: 10.3390/ijms20225771] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/04/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic and relapsing immune disorders that result, or possibly originate, from epithelial barrier defects. Intestinal organoids are a new reliable tool to investigate epithelial response in models of chronic inflammation. We produced organoids from the ulcerative colitis murine model Winnie to explore if the chronic inflammatory features observed in the parental intestine were preserved by the organoids. Furthermore, we investigated if quercetin administration to in vitro cultured organoids could suppress LPS-induced inflammation in wild-type organoids (WT-organoids) and spontaneous inflammation in ulcerative colitis organoids (UC-organoids). Our data demonstrate that small intestinal organoids obtained from Winnie mice retain the chronic intestinal inflammatory features characteristic of the parental tissue. Quercetin administration was able to suppress inflammation both in UC-organoids and in LPS-treated WT-organoids. Altogether, our data demonstrate that UC-organoids are a reliable experimental system for investigating chronic intestinal inflammation and pharmacological responses.
Collapse
Affiliation(s)
- Manuela Dicarlo
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
- Correspondence: (M.D.); (M.C.); Tel.: +39-089-233463 (M.C.)
| | - Gabriella Teti
- Department of Biomedical and Neuromotor Sciences-DBNS, Università di Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.T.); (M.F.)
| | - Giulio Verna
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Marina Liso
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Elisabetta Cavalcanti
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Annamaria Sila
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Sathuwarman Raveenthiraraj
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (S.R.); (A.S.)
| | - Mauro Mastronardi
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Angelo Santino
- ISPA-CNR, Institute of Science of Food Production, C.N.R. Unit of Lecce, 73100 Lecce, Italy;
| | - Grazia Serino
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Antonio Lippolis
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
| | - Anastasia Sobolewski
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK; (S.R.); (A.S.)
| | - Mirella Falconi
- Department of Biomedical and Neuromotor Sciences-DBNS, Università di Bologna, Via Irnerio 48, 40126 Bologna, Italy; (G.T.); (M.F.)
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte (BA), Italy; (G.V.); (M.L.); (E.C.); (A.S.); (M.M.); (G.S.); (A.L.)
- Department of Immunology and Cell Biology, European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy
- Correspondence: (M.D.); (M.C.); Tel.: +39-089-233463 (M.C.)
| |
Collapse
|
21
|
Yin YB, de Jonge HR, Wu X, Yin YL. Mini-gut: a promising model for drug development. Drug Discov Today 2019; 24:1784-1794. [PMID: 31212027 DOI: 10.1016/j.drudis.2019.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/01/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023]
Abstract
Until recently, major advances in drug development have been hampered by a lack of proper cell and tissue models; but the introduction of organoid technology has revolutionized this field. At the level of the gastrointestinal tract, the so-called mini-gut comprises all major cell types of native intestine and recapitulates the composition and function of native intestinal epithelium. The mini-gut can be classified as an intestinal organoid (IO), derived from pluripotent stem cells, or as an enteroid, consisting only of epithelial cells and generated from adult stem cells. Both classifications have been used as models to develop drugs against cystic fibrosis, cancer and infectious disease, as well as for drug screening, personalized medicine and the development of new medical tools. In this review, we highlight and discuss the importance of mini-guts for drug development and point out their limitations and future prospects.
Collapse
Affiliation(s)
- Yue-Bang Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan, 410125, China; Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Xin Wu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan, 410125, China; Institute of Biological Resources, Jiangxi Academy of Sciences, Nanchang 330096, China.
| | - Yu-Long Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, Hunan, 410125, China.
| |
Collapse
|