1
|
Tanaka K, Kawai S, Fujii E, Yano M, Miyayama T, Nakano K, Terao K, Suzuki M. Development of rat duodenal monolayer model with effective barrier function from rat organoids for ADME assay. Sci Rep 2023; 13:12130. [PMID: 37495742 PMCID: PMC10372144 DOI: 10.1038/s41598-023-39425-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023] Open
Abstract
The in-depth analysis of the ADME profiles of drug candidates using in vitro models is essential for drug development since a drug's exposure in humans depends on its ADME properties. In contrast to efforts in developing human in vitro absorption models, only a limited number of studies have explored models using rats, the most frequently used species in in vivo DMPK studies. In this study, we developed a monolayer model with an effective barrier function for ADME assays using rat duodenal organoids as a cell source. At first, we developed rat duodenal organoids according to a previous report, but they were not able to generate a confluent monolayer. Therefore, we modified organoid culture protocols and developed cyst-enriched organoids; these strongly promoted the formation of a confluent monolayer. Furthermore, adding valproic acid to the culture accelerated the differentiation of the monolayer, which possessed an effective barrier function and apicobasal cell polarity. Drug transporter P-gp function as well as CYP3A activity and nuclear receptor function were confirmed in the model. We expect our novel monolayer model to be a useful tool for elucidating drug absorption processes in detail, enabling the development of highly absorbable drugs.
Collapse
Affiliation(s)
- Kai Tanaka
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 5-1-1 Tsukiji Chuo-Ku, Tokyo, 104-0045, Japan.
| | - Shigeto Kawai
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 5-1-1 Tsukiji Chuo-Ku, Tokyo, 104-0045, Japan
| | - Etsuko Fujii
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka Totsuka-Ku Yokohama, Kanagawa, 244-8602, Japan
| | - Masumi Yano
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka Totsuka-Ku Yokohama, Kanagawa, 244-8602, Japan
| | - Takashi Miyayama
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka Totsuka-Ku Yokohama, Kanagawa, 244-8602, Japan
| | - Kiyotaka Nakano
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 5-1-1 Tsukiji Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kimio Terao
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 2-1-1 Nihonbashi-Muromachi Chuo-Ku, Tokyo, 103-8324, Japan
| | - Masami Suzuki
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| |
Collapse
|
2
|
Qin Y, Wang S, Huang W, Li K, Wu M, Liu W, Han J. Chlorogenic acid improves intestinal morphology by enhancing intestinal stem-cell activity. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:3287-3294. [PMID: 36698257 DOI: 10.1002/jsfa.12469] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 12/11/2022] [Accepted: 01/26/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Chlorogenic acid (CGA), as one of the most abundant naturally occurring phenolic acids, has been documented to be beneficial for intestinal health. However, the underlying mechanism is still not fully understood. The adult intestinal stem cell is the critical driver of epithelial homeostasis and regeneration. RESULTS This study hypothesized that CGA exerted intestinal health effects by modulating intestinal stem-cell functions. Lgr5-EGFP mice were treated for 14 days, and intestinal organoids derived from these mice were treated for 3 days, using CGA solution. In comparison with the control group, CGA treatment increased intestinal villous height and crypt depth in mice and augmented the area expansion and the number of budding intestinal organoids. Quantitative polymerase chain reaction (qPCR) analysis revealed that CGA treatment significantly increased the expression of genes coding intestinal stem-cell markers in intestinal tissue and organoids, and upregulated the expression of genes coding secretory cell lineages and enterocytes, although not statistically significantly. Fluorescence-activated cell-sorting analysis further confirmed that CGA augmented the number of stem cells. 5-Ethynyl-2'-deoxyuridine (EdU) incorporation and Ki67 immunostaining results also demonstrated that CGA treatment enhanced intestinal stem-cell proliferation. CONCLUSION Altogether, our findings indicate that CGA could activate intestinal stem-cell and epithelial regeneration, which could contribute to the improvement of intestinal morphology or organoid growth of mice. This highlights a promising mechanism for CGA as an excellent candidate for the formulation of dietary supplements and functional foods for intestinal protection. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yumei Qin
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Suqiang Wang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Weiwei Huang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Kejin Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Min Wu
- Ecology and Health Institute, Hangzhou Vocational and Technical College, Hangzhou, China
| | - Weilin Liu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Jianzhong Han
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| |
Collapse
|
3
|
Doyle ME, Premathilake HU, Yao Q, Mazucanti CH, Egan JM. Physiology of the tongue with emphasis on taste transduction. Physiol Rev 2023; 103:1193-1246. [PMID: 36422992 PMCID: PMC9942923 DOI: 10.1152/physrev.00012.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The tongue is a complex multifunctional organ that interacts and senses both interoceptively and exteroceptively. Although it is easily visible to almost all of us, it is relatively understudied and what is in the literature is often contradictory or is not comprehensively reported. The tongue is both a motor and a sensory organ: motor in that it is required for speech and mastication, and sensory in that it receives information to be relayed to the central nervous system pertaining to the safety and quality of the contents of the oral cavity. Additionally, the tongue and its taste apparatus form part of an innate immune surveillance system. For example, loss or alteration in taste perception can be an early indication of infection as became evident during the present global SARS-CoV-2 pandemic. Here, we particularly emphasize the latest updates in the mechanisms of taste perception, taste bud formation and adult taste bud renewal, and the presence and effects of hormones on taste perception, review the understudied lingual immune system with specific reference to SARS-CoV-2, discuss nascent work on tongue microbiome, as well as address the effect of systemic disease on tongue structure and function, especially in relation to taste.
Collapse
Affiliation(s)
- Máire E Doyle
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Hasitha U Premathilake
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Qin Yao
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Caio H Mazucanti
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M Egan
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
4
|
Sénicourt B, Cloutier G, Basora N, Fallah S, Laniel A, Lavoie C, Beaulieu JF. Primary Cilium Identifies a Quiescent Cell Population in the Human Intestinal Crypt. Cells 2023; 12:cells12071059. [PMID: 37048132 PMCID: PMC10093653 DOI: 10.3390/cells12071059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Primary cilia are sensory antennae located at the cell surface which mediate a variety of extracellular signals involved in development, tissue homeostasis, stem cells and cancer. Primary cilia are found in an extensive array of vertebrae cells but can only be generated when cells become quiescent. The small intestinal epithelium is a rapidly self-renewing tissue organized into a functional unit called the crypt–villus axis, containing progenitor and differentiated cells, respectively. Terminally differentiated villus cells are notoriously devoid of primary cilia. We sought to determine if intestinal crypts contain a quiescent cell population that could be identified by the presence of primary cilia. Here we show that primary cilia are detected in a subset of cells located deep in the crypts slightly above a Paneth cell population. Using a normal epithelial proliferative crypt cell model, we show that primary cilia assembly and activity correlate with a quiescent state. These results provide further evidence for the existence of a quiescent cell population in the human small intestine and suggest the potential for new modes of regulation in stem cell dynamics.
Collapse
|
5
|
Zhou JY, Zan GX, Zhu QJ, Gao CQ, Yan HC, Wang XQ. Recombinant Porcine R-Spondin 1 Facilitates Intestinal Stem Cell Expansion along the Crypt-Villus Axis through Potentiating Wnt/β-Catenin Signaling in Homeostasis and Deoxynivalenol Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10644-10653. [PMID: 35997221 DOI: 10.1021/acs.jafc.2c02013] [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: 06/15/2023]
Abstract
R-spondin 1 (RSPO1) is a ligand for the intestinal stem cell (ISC) marker Lgr5 in the crypt, which functions to amplify canonical Wnt signaling to stimulate the division of ISCs. Despite the crucial role of recombinant human RSPO1 (rhRSPO1) in homeostasis and regeneration, little is known about RSPO1 among different species. Here, we cloned the porcine RSPO1 (pRSPO1) gene and obtained rpRSPO1 protein through the expression system of the recombinant Escherichia coli Rosetta (DE3) chemical competent cells. Using the in vitro IPEC-J2 model that combines cell proliferation evaluation approaches, we identified the rpRSPO1 activity in stimulating jejunal epithelial cells. And upon deoxynivalenol challenge in mice, we found that rpRSPO1 ameliorated their growth retardation and jejunal epithelial integrity. Importantly, the ISCs in the jejunum had greater proliferation and differentiation potential that was accompanied by Wnt/β-catenin pathway activation after rpRSPO1 modulation. Subsequently, the jejunal organoids expanded from these ISCs ex vivo presented robust growth advantages. And the rpRSPO1 was able to guide Wnt/β-catenin activity to increase ISC activity. Our work systematically demonstrates that rpRSPO1 facilitates ISC expansion by potentiating Wnt/β-catenin signaling during homeostasis and responding to deoxynivalenol perturbations.
Collapse
Affiliation(s)
- Jia-Yi Zhou
- Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- HenryFok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China
| | - Geng-Xiu Zan
- Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qiu-Jie Zhu
- Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Chun-Qi Gao
- Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hui-Chao Yan
- Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiu-Qi Wang
- Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
6
|
Zhou J, Hou C, Chen H, Qin Z, Miao Z, Zhao J, Wang Q, Cui M, Xie C, Wang R, Li Q, Zuo G, Miao D, Jin J. P16 I NK 4a Deletion Ameliorates Damage of Intestinal Epithelial Barrier and Microbial Dysbiosis in a Stress-Induced Premature Senescence Model of Bmi-1 Deficiency. Front Cell Dev Biol 2021; 9:671564. [PMID: 34712655 PMCID: PMC8545785 DOI: 10.3389/fcell.2021.671564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
This study aimed to determine whether Bmi-1 deficiency leads to intestinal epithelial barrier destruction and microbiota dysfunction, which members of the microbial community alter barrier function with age, and whether p16INK4a deletion could reverse the damage of intestinal epithelial barrier and microbial dysbiosis. Intestines from Bmi-1–deficient (Bmi-1–/–), Bmi-1 and p16INK4a double-knockout (Bmi-1–/–p16INK4a–/–), and wild-type mice were observed for aging and inflammation. Duolink Proximity Ligation Assay, immunoprecipitation, and construction of p16INK4a overexpressed adenovirus and the overexpressed plasmids of full-length, mutant, or truncated fragments for occludin were used for analyzing the interaction between p16INK4a and occludin. High-throughput sequencing of V4 region amplicon of 16S ribosomal RNA was conducted using intestinal microbiota. We found Bmi-1 deficiency destructed barrier structure, barrier function, and tight junction (TJ) in intestinal epithelium; decreased the TJ proteins; increased tumor necrosis factor α (TNF-α)–dependent barrier permeability; and up-regulated proinflammatory level of macrophages induced by intestinal microbial dysbiosis. The transplantation of fecal microbiota from wild-type mice ameliorated TJ in intestinal epithelium of Bmi-1–/– and Bmi-1–/–p16INK4a–/– mice. Harmful bacteria including Desulfovibrio, Helicobacter, and Oscillibacter were at a higher level in Bmi-1–/– mice. More harmful bacteria Desulfovibrio entered the epithelium and promoted macrophages-secreted TNF-α and caused TNF-α–dependent barrier permeability and aging. Accumulated p16INK4a combined with occludin at the 1st–160th residue in cytoplasm of intestinal epithelium cells from Bmi-1–/– mice, which blocked formation of TJ and the repair of intestinal epithelium barrier. P16INK4a deletion could maintain barrier function and microbiota balance in Bmi-1–/– mice through strengthening formation of TJ and decreasing macrophages-secreted TNF-α induced by Desulfovibrio entering the intestinal epithelium. Thus, Bmi-1 maintained intestinal TJ, epithelial barrier function, and microbiota balance through preventing senescence characterized by p16INK4a accumulation. The clearance of p16INK4a-positive cells in aging intestinal epithelium would be a new method for maintaining barrier function and microbiota balance. The residues 1–160 of occludin could be a novel therapeutic target for identifying small molecular antagonistic peptides to prevent the combination of p16INK4a with occludin for protecting TJ.
Collapse
Affiliation(s)
- Jiawen Zhou
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chenxing Hou
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Haiyun Chen
- Anti-Aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Ziyue Qin
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Zi'an Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jingyu Zhao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qiuyi Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Min Cui
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rong Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, China
| | - Guoping Zuo
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Anti-Aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Jianliang Jin
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Cortés-Llanos B, Wang Y, Sims CE, Allbritton NL. A technology of a different sort: microraft arrays. LAB ON A CHIP 2021; 21:3204-3218. [PMID: 34346456 PMCID: PMC8387436 DOI: 10.1039/d1lc00506e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A common procedure performed throughout biomedical research is the selection and isolation of biological entities such as organelles, cells and organoids from a mixed population. In this review, we describe the development and application of microraft arrays, an analysis and isolation platform which enables a vast range of criteria and strategies to be used when separating biological entities. The microraft arrays are comprised of elastomeric microwells with detachable polymer bases (microrafts) that act as capture and culture sites as well as supporting carriers during cell isolation. The technology is elegant in its simplicity and can be implemented for samples possessing tens to millions of objects yielding a flexible platform for applications such as single-cell RNA sequencing, subcellular organelle capture and assay, high-throughput screening and development of CRISPR gene-edited cell lines, and organoid manipulation and selection. The transparent arrays are compatible with a multitude of imaging modalities enabling selection based on 2D or 3D spatial phenotypes or temporal properties. Each microraft can be individually isolated on demand with retention of high viability due to the near zero hydrodynamic stress imposed upon the cells during microraft release, capture and deposition. The platform has been utilized as a simple manual add-on to a standard microscope or incorporated into fully automated instruments that implement state-of-the-art imaging algorithms and machine learning. The vast array of selection criteria enables separations not possible with conventional sorting methods, thus garnering widespread interest in the biological and pharmaceutical sciences.
Collapse
|
8
|
Ftuwi H, Parri R, Mohammed AR. Novel, Fully Characterised Bovine Taste Bud Cells of Fungiform Papillae. Cells 2021; 10:2285. [PMID: 34571933 PMCID: PMC8469975 DOI: 10.3390/cells10092285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/23/2021] [Accepted: 08/28/2021] [Indexed: 01/14/2023] Open
Abstract
Current understanding of functional characteristics and biochemical pathways in taste bud cells have been hindered due the lack of long-term cultured cells. To address this, we developed a holistic approach to fully characterise long term cultured bovine taste bud cells (BTBCs). Initially, cultured BTBCs were characterised using RT-PCR gene expression profiling, immunocytochemistry, flowcytometry and calcium imaging, that confirmed the cells were mature TBCs that express taste receptor genes, taste specific protein markers and capable of responding to taste stimuli, i.e., denatonium (2 mM) and quinine (462.30 μM). Gene expression analysis of forty-two genes implicated in taste transduction pathway (map04742) using custom-made RT-qPCR array revealed high and low expressed genes in BTBCs. Preliminary datamining and bioinformatics demonstrated that the bovine α-gustducin, gustatory G-protein, have higher sequence similarity to the human orthologue compared to rodents. Therefore, results from this work will replace animal experimentation and provide surrogate cell-based throughput system to study human taste transduction.
Collapse
Affiliation(s)
| | | | - Afzal R. Mohammed
- Aston Pharmacy School, Aston University, Birmingham B4 7ET, UK; (H.F.); (R.P.)
| |
Collapse
|
9
|
Zhou JY, Huang DG, Gao CQ, Yan HC, Zou SG, Wang XQ. Heat-stable enterotoxin inhibits intestinal stem cell expansion to disrupt the intestinal integrity by downregulating the Wnt/β-catenin pathway. Stem Cells 2021; 39:482-496. [PMID: 33373490 DOI: 10.1002/stem.3324] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Enterotoxigenic Escherichia coli causes severe infectious diarrhea with high morbidity and mortality in newborn and weanling pigs mainly through the production of heat-stable enterotoxins (STs). However, the precise regulatory mechanisms involved in ST-induced intestinal epithelium injury remain unclear. Consequently, we conducted the experiments in vivo (mice), ex vivo (mouse and porcine enteroids), and in vitro (MODE-K and IPEC-J2 cells) to explore the effect of STp (one type of STa) on the integrity of the intestinal epithelium. The results showed that acute STp exposure led to small intestinal edema, disrupted intestinal integrity, induced crypt cell expansion into spheroids, and downregulated Wnt/β-catenin activity in the mice. Following a similar trend, the enteroid-budding efficiency and the expression of Active β-catenin, β-catenin, Lgr5, PCNA, and KRT20 were significantly decreased after STp treatment, as determined ex vivo. In addition, STp inhibited cell proliferation, induced cell apoptosis, destroyed cell barriers, and reduced Wnt/β-catenin activity by downregulating its membrane receptor Frizzled7 (FZD7). In contrast, Wnt/β-catenin reactivation protected the IPEC-J2 cells from STp-induced injury. Taking these findings together, we conclude that STp inhibits intestinal stem cell expansion to disrupt the integrity of the intestinal mucosa through the downregulation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jia-Yi Zhou
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, People's Republic of China
| | - Deng-Gui Huang
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, People's Republic of China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, People's Republic of China.,Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, People's Republic of China
| | - Hui-Chao Yan
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, People's Republic of China
| | - Shi-Geng Zou
- Wen's Group Academy, Wen's Foodstuffs Group Co, Ltd, Xinxing, People's Republic of China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, People's Republic of China
| |
Collapse
|
10
|
Signaling Network Centered on mTORC1 Dominates Mammalian Intestinal Stem Cell Ageing. Stem Cell Rev Rep 2020; 17:842-849. [PMID: 33201440 DOI: 10.1007/s12015-020-10073-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 12/19/2022]
Abstract
The intestine integrates the function of digestion, absorption, and barrier, which is easily damaged by the external factors upon ageing. The intestinal stem cells (ISCs) exist at the intestinal crypt base and play an indispensable role in intestinal homeostasis and regeneration. The intestine ageing contributes to malabsorption and other associated illnesses, which were considered to be related to ISCs. Here, we summarize the current research progress of mammalian ISCs ageing and pay more attention to the central regulatory role of the mTORC1 signaling pathway in regulating mammalian ISCs ageing, and its related AMPK, FOXO, Wnt signaling pathways. Furthermore, we also discuss the interventions aimed at mTORC1 and its associated signaling pathways, which may provide potential strategies for rejuvenating aged ISCs and the therapy of age-related intestinal diseases. Graphical abstract Many signaling pathways are altered in the ageing ISCs, thereby inducing the decrease of ISC self-renewal, differentiation, and regeneration, an increasing of oxidative stress may contribute to damage to the ISCs. Interventions such as calorie restriction, fasting and so on can effectively alleviate these adverse effects.
Collapse
|
11
|
Walker BS, Zarour LR, Wieghard N, Gallagher AC, Swain JR, Weinmann S, Lanciault C, Billingsley K, Tsikitis VL, Wong MH. Stem Cell Marker Expression in Early Stage Colorectal Cancer is Associated with Recurrent Intestinal Neoplasia. World J Surg 2020; 44:3501-3509. [PMID: 32647988 PMCID: PMC10659815 DOI: 10.1007/s00268-020-05586-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) ranks second in cancer deaths worldwide and presents multiple management challenges, one of which is identifying high risk stage II disease that may benefit from adjuvant therapy. Molecular biomarkers, such as ones that identify stem cell activity, could better stratify high-risk cohorts for additional treatment. METHODS To identify possible biomarkers of high-risk disease in early-stage CRC, a discovery set (n = 66) of advanced-stage tumors were immunostained with antibodies to stemness proteins (CD166, CD44, CD26, and LGR5) and then digitally analyzed. Using a second validation cohort (n = 54) of primary CRC tumors, we analyzed protein and gene expression of CD166 across disease stages, and extended our analyses to CD166-associated genes (LGR5, ASCL2, BMI1, POSTN, and VIM) by qRT-PCR. RESULTS Stage III and metastatic CRC tumors highly expressed stem cell-associated proteins, CD166, CD44, and LGR5. When evaluated across stages, CD166 protein expression was elevated in advanced-stage compared to early-stage tumors. Notably, a small subset of stage I and II cancers harbored elevated CD166 protein expression, which correlated with development of recurrent cancer or adenomatous polyps. Gene expression analyses of CD166-associated molecules revealed elevated ASCL2 in primary tumors from patients who recurred. CONCLUSIONS We identified a protein signature prognostic of aggressive disease in early stage CRC. Stem cell-associated protein and gene expression identified a subset of early-stage tumors associated with cancer recurrence and/or subsequent adenoma formation. Signatures for stemness offer promising fingerprints for stratifying early-stage patients at high risk of recurrence.
Collapse
Affiliation(s)
- Brett S Walker
- Department of Surgery, OHSU, 3181 SW Sam Jackson Park Rd, L619, Portland, OR, 97239, USA
| | - Luai R Zarour
- Department of Surgery, OHSU, 3181 SW Sam Jackson Park Rd, L619, Portland, OR, 97239, USA
| | - Nicole Wieghard
- Department of Surgery, OHSU, 3181 SW Sam Jackson Park Rd, L619, Portland, OR, 97239, USA
| | - Alexandra C Gallagher
- Department of Cell, Developmental, and Cancer Biology, OHSU, 2720 S Moody Ave., KR-CDCB, Portland, OR, 97201, USA
| | - John R Swain
- Department of Cell, Developmental, and Cancer Biology, OHSU, 2720 S Moody Ave., KR-CDCB, Portland, OR, 97201, USA
| | - Sheila Weinmann
- Kaiser Permanente Northwest Center for Health Research, 3800 N. Interstate Ave., Portland, OR, 97227, USA
| | - Christian Lanciault
- Department of Pathology, OHSU, 3181 SW Sam Jackson Park Rd, L-113, Portland, OR, 97239, USA
| | - Kevin Billingsley
- Department of Surgery, OHSU, 3181 SW Sam Jackson Park Rd, L619, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Ave., Portland, OR, 97201, USA
| | - V Liana Tsikitis
- Department of Surgery, OHSU, 3181 SW Sam Jackson Park Rd, L619, Portland, OR, 97239, USA.
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Ave., Portland, OR, 97201, USA.
| | - Melissa H Wong
- Department of Cell, Developmental, and Cancer Biology, OHSU, 2720 S Moody Ave., KR-CDCB, Portland, OR, 97201, USA.
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Ave., Portland, OR, 97201, USA.
| |
Collapse
|
12
|
Yoon JY, Brezden-Masley C, Streutker CJ. Lgr5 and stem/progenitor gene expression in gastric/gastroesophageal junction carcinoma - significance of potentially retained stemness. BMC Cancer 2020; 20:860. [PMID: 32894084 PMCID: PMC7487651 DOI: 10.1186/s12885-020-07362-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background Gastric/gastroesophageal junction (GEJ) adenocarcinomas are heterogeneous, comprising four molecularly distinct subtypes, namely EBV-positive, microsatellite instability (MSI), chromosomal instability (CIN) and genomically stable (GS) subtypes, and a part of this heterogeneity may hypothesized to be different cells-of-origin. Stem/progenitor cell hierarchy in the stomach is complex, which include the Lgr5(+) gastric stem cells (GSCs). Methods While previous studies have focused on non-nuclear Lgr5 expression, nuclear Lgr5 expression has been reported in a subset of stem cells, and we examined nuclear Lgr5 expression in a local cohort of 95 cases of gastric/GEJ adenocarcinoma. mRNA levels for LGR5 and other stem cell marker genes were examined in the TCGA cohort. Results We observed nuclear Lgr5 expression in a 18/95 cases. Near mutual exclusivity was seen between nuclear Lgr5 and strong non-nuclear Lgr5. Both strong non-nuclear and nuclear Lgr5 expression tended to be seen more frequently with the intestinal histotype and approximated CIN molecular subtype. With respect to overall survival (OS), nuclear Lgr5 expression appears to be protective, with the worst survival being seen in the cases lacking nuclear Lgr5 and with low non-nuclear Lgr5 expression. When compared to other stem/progenitor cell markers, LGR5 mRNA expression clusters with other GSC marker genes, including VIL1. Higher expression of these GSC marker genes was associated with better OS. Conclusions Our results show that Lgr5 expression is dynamic in gastric/GEJ adenocarcinoma and heterogeneous across the several disease attributes. We postulate that this may reflect “retained stemness” in the form of Lgr5High-GSC signature that appears to be associated with better survival.
Collapse
Affiliation(s)
- Ju-Yoon Yoon
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Catherine J Streutker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. .,Department of Pathology, St. Michael's Hospital, St. Michael's Hospital, Unity Health Toronto, Rm 2-099CC, 30 Bond Street, Toronto, Ontario, M5B-1W8, Canada.
| |
Collapse
|
13
|
Min J, Vega PN, Engevik AC, Williams JA, Yang Q, Patterson LM, Simmons AJ, Bliton RJ, Betts JW, Lau KS, Magness ST, Goldenring JR, Choi E. Heterogeneity and dynamics of active Kras-induced dysplastic lineages from mouse corpus stomach. Nat Commun 2019; 10:5549. [PMID: 31804471 PMCID: PMC6895174 DOI: 10.1038/s41467-019-13479-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Dysplasia is considered a key transition state between pre-cancer and cancer in gastric carcinogenesis. However, the cellular or phenotypic heterogeneity and mechanisms of dysplasia progression have not been elucidated. We have established metaplastic and dysplastic organoid lines, derived from Mist1-Kras(G12D) mouse stomach corpus and studied distinct cellular behaviors and characteristics of metaplastic and dysplastic organoids. We also examined functional roles for Kras activation in dysplasia progression using Selumetinib, a MEK inhibitor, which is a downstream mediator of Kras signaling. Here, we report that dysplastic organoids die or show altered cellular behaviors and diminished aggressive behavior in response to MEK inhibition. However, the organoids surviving after MEK inhibition maintain cellular heterogeneity. Two dysplastic stem cell (DSC) populations are also identified in dysplastic cells, which exhibited different clonogenic potentials. Therefore, Kras activation controls cellular dynamics and progression to dysplasia, and DSCs might contribute to cellular heterogeneity in dysplastic cell lineages.
Collapse
Affiliation(s)
- Jimin Min
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Paige N Vega
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Janice A Williams
- Cell Imaging Share Resource, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Qing Yang
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Institute of Pathogen Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Loraine M Patterson
- Center for GI Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alan J Simmons
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - R Jarrett Bliton
- UNC Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joshua W Betts
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Scott T Magness
- Center for GI Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- University of North Carolina Chapel Hill/ North Carolina State University joint Departments of Biomedical Engineering, Chapel Hill, NC, 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Nashville VA Medical Center, Nashville, TN, 37232, USA
| | - Eunyoung Choi
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| |
Collapse
|
14
|
Effect of Antifreeze Glycoproteins on Organoid Survival during and after Hypothermic Storage. Biomolecules 2019; 9:biom9030110. [PMID: 30893938 PMCID: PMC6468685 DOI: 10.3390/biom9030110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/08/2019] [Accepted: 03/15/2019] [Indexed: 11/23/2022] Open
Abstract
We study the effect of antifreeze glycoproteins (AFGPs) on the survival of organoids under hypothermic conditions. We find that the survival of organoids in cold conditions depends on their developmental stage. Mature organoids die within 24 h when being stored at 4 °C, while cystic organoids can survive up to 48 h. We find that in the presence of AFGPs, the organoid survival is prolonged up to 72 h, irrespective of their developmental stage. Fluorescence microscopy experiments reveal that the AFGPs predominately localize at the cell surface and cover the cell membranes. Our findings support a mechanism in which the positive effect of AFGPs on cell survival during hypothermic storage involves the direct interaction of AFGPs with the cell membrane. Our research highlights organoids as an attractive multicellular model system for studying the action of AFGPs that bridges the gap between single-cell and whole-organ studies.
Collapse
|
15
|
Sheahan B, Dekaney CM. Decrypting the Crypt: Novel Monoclonal Antibodies to Identify Intestinal Stem Cell Populations. Cell Mol Gastroenterol Hepatol 2018; 6:121-122. [PMID: 29928681 PMCID: PMC6007819 DOI: 10.1016/j.jcmgh.2018.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
| | - Christopher M. Dekaney
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|