1
|
Liu H, Yang G, Wang H. Oxytocin/Oxytocin Receptor Signalling in the Gastrointestinal System: Mechanisms and Therapeutic Potential. Int J Mol Sci 2024; 25:10935. [PMID: 39456718 PMCID: PMC11508134 DOI: 10.3390/ijms252010935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The neuropeptide hormone oxytocin (OT) is involved in various physiological and pathological processes via the oxytocin receptor (OTR). While OT is most widely known as a reproductive system hormone and a nervous system neurotransmitter, the OT/OTR system has gradually gained much attention for its role in the gastrointestinal (GI) system, such as the GI motility, secretion, and bowel inflammatory reactions. Its importance in GI cancers has also been reported in the past few decades. The promising clinical observations have revealed OT's anti-nociceptive effect, protective effect over gut injury, and the potential of using microbiota to naturally increase endogenous OT levels, which shed a light on the management of GI disorders with lower side effects. However, no current comprehensive review is available on the actions of OT/OTR in the GI tract. This review aims to present the lesser-known role of the OT/OTR system in the GI tract, and the most recent findings are discussed regarding the distribution and functional role of OTR signalling in regulating (patho)physiological functions of the GI tract. Special emphasis is placed on its therapeutic potential for clinical management of GI disorders, such as GI pain, inflammatory bowel disease (IBD), and irritable bowel syndrome (IBS). The recent characterisation of the OTR's crystal structure has advanced research for designing and identifying new OTR-specific molecules. Future in-depth basic and clinical research is needed to further elucidate the involvement and detailed mechanism of OT/OTR in GI disorders, and the development of OTR-specific ligands.
Collapse
Affiliation(s)
- Huiping Liu
- School of Pharmacy, Yantai University, Yantai 264005, China; (G.Y.); (H.W.)
| | | | | |
Collapse
|
2
|
Tsuchida A, Hachisu K, Mizuno M, Takada Y, Ideo H. High expression of B3GALT5 suppresses the galectin-4-mediated peritoneal dissemination of poorly differentiated gastric cancer cells. Glycobiology 2024; 34:cwae064. [PMID: 39163480 DOI: 10.1093/glycob/cwae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
Peritoneal metastasis frequently accompanies metastatic and/or recurrent gastric cancer, leading to a poor prognosis owing to a lack of effective treatment. Hence, there is a pressing need to enhance our understanding of the mechanisms and molecules driving peritoneal metastasis. In a previous study, galectin-4 inhibition impeded peritoneal metastasis in a murine model. This study examined the glycan profiles of cell surface proteins and glycosphingolipids (GSLs) in cells with varying tumorigenic potentials to understand the intricate mechanisms underlying galectin-4-mediated regulation, particularly glycosylation. Detailed mass spectrometry analysis showed that galectin-4 knockout cells exhibit increased expression of lacto-series GSLs with β1,3-linked galactose while showing no significant alterations in neolacto-series GSLs. We conducted real-time polymerase chain reaction (PCR) analysis to identify candidate glycosyltransferases that synthesize increased levels of GSLs. Subsequently, we introduced the candidate B3GALT5 gene and selected the clones with high expression levels. B3GALT5 gene-expressing clones showed GSL glycan profiles like those of knockout cells and significantly reduced tumorigenic ability in mouse models. These clones exhibited diminished proliferative capacity and showed reduced expression of galectin-4 and activated AKT. Moreover, co-localization of galectin-4 with flotillin-2 (a raft marker) decreased in B3GALT5-expressing cells, implicating GSLs in galectin-4 localization to lipid rafts. D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (a GSL synthase inhibitor) also affected galectin-4 localization in rafts, suggesting the involvement of GSL microdomains. We discovered that B3GALT5 plays a crucial role in regulating peritoneal metastasis of malignant gastric cancer cells by suppressing cell proliferation and modulating lipid rafts and galectin-4 via mechanisms that are yet to be elucidated.
Collapse
Affiliation(s)
- Akiko Tsuchida
- Laboratory of Glycobiology, The Noguchi Institute, 1-9-7, Kaga, Itabashi, Tokyo 173-0003, Japan
| | - Kazuko Hachisu
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-9-7, Kaga, Itabashi, Tokyo 173-0003, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-9-7, Kaga, Itabashi, Tokyo 173-0003, Japan
| | - Yoshio Takada
- Laboratory of Glycobiology, The Noguchi Institute, 1-9-7, Kaga, Itabashi, Tokyo 173-0003, Japan
| | - Hiroko Ideo
- Laboratory of Glycobiology, The Noguchi Institute, 1-9-7, Kaga, Itabashi, Tokyo 173-0003, Japan
| |
Collapse
|
3
|
Sachan N, Sharma V, Mutsuddi M, Mukherjee A. Notch signalling: multifaceted role in development and disease. FEBS J 2024; 291:3030-3059. [PMID: 37166442 DOI: 10.1111/febs.16815] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Notch pathway is an evolutionarily conserved signalling system that operates to influence an astonishing array of cell fate decisions in different developmental contexts. Notch signalling plays important roles in many developmental processes, making it difficult to name a tissue or a developing organ that does not depend on Notch function at one stage or another. Thus, dysregulation of Notch signalling is associated with many developmental defects and various pathological conditions, including cancer. Although many recent advances have been made to reveal different aspects of the Notch signalling mechanism and its intricate regulation, there are still many unanswered questions related to how the Notch signalling pathway functions in so many developmental events. The same pathway can be deployed in numerous cellular contexts to play varied and critical roles in an organism's development and this is only possible because of the complex regulatory mechanisms of the pathway. In this review, we provide an overview of the mechanism and regulation of the Notch signalling pathway along with its multifaceted functions in different aspects of development and disease.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
4
|
Kim JE, Tun HM, Bennett DC, Leung FC, Cheng KM. Microbial diversity and metabolic function in duodenum, jejunum and ileum of emu (Dromaius novaehollandiae). Sci Rep 2023; 13:4488. [PMID: 36934111 PMCID: PMC10024708 DOI: 10.1038/s41598-023-31684-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 03/15/2023] [Indexed: 03/20/2023] Open
Abstract
Emus (Dromaius novaehollandiae), a large flightless omnivorous ratite, are farmed for their fat and meat. Emu fat can be rendered into oil for therapeutic and cosmetic use. They are capable of gaining a significant portion of its daily energy requirement from the digestion of plant fibre. Despite of its large body size and low metabolic rate, emus have a relatively simple gastroinstetinal (GI) tract with a short mean digesta retention time. However, little is known about the GI microbial diversity of emus. The objective of this study was to characterize the intraluminal intestinal bacterial community in the different segments of small intestine (duodenum, jejunum, and ileum) using pyrotag sequencing and compare that with the ceca. Gut content samples were collected from each of four adult emus (2 males, 2 females; 5-6 years old) that were free ranged but supplemented with a barley-alfalfa-canola based diet. We amplified the V3-V5 region of 16S rRNA gene to identify the bacterial community using Roche 454 Junior system. After quality trimming, a total of 165,585 sequence reads were obtained from different segments of the small intestine (SI). A total of 701 operational taxonomic units (OTUs) were identified in the different segments of small intestine. Firmicutes (14-99%) and Proteobacteria (0.5-76%) were the most predominant bacterial phyla in the small intestine. Based on species richness estimation (Chao1 index), the average number of estimated OTUs in the small intestinal compartments were 148 in Duodenum, 167 in Jejunum, and 85 in Ileum, respectively. Low number of core OTUs identified in each compartment of small intestine across individual birds (Duodenum: 13 OTUs, Jejunum: 2 OTUs, Ileum: 14 OTUs) indicated unique bacterial community in each bird. Moreover, only 2 OTUs (Escherichia and Sinobacteraceae) were identified as core bacteria along the whole small intestine. PICRUSt analysis has indicated that the detoxification of plant material and environmental chemicals seem to be performed by SI microbiota, especially those in the jejunum. The emu cecal microbiome has more genes than SI segments involving in protective or immune response to enteric pathogens. Microbial digestion and fermentation is mostly in the jejunum and ceca. This is the first study to characterize the microbiota of different compartments of the emu intestines via gut samples and not fecal samples. Results from this study allow us to further investigate the influence of the seasonal and physiological changes of intestinal microbiota on the nutrition of emus and indirectly influence the fatty acid composition of emu fat.
Collapse
Affiliation(s)
- Ji Eun Kim
- Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Hein M Tun
- School of Public Health, Li Ka Shing, Faculty of Medicine, HKU-Pasteur Research Pole, University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
- JC School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Darin C Bennett
- Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Frederick C Leung
- School of Biological Sciences, Faculty of Science, University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Kimberly M Cheng
- Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada.
| |
Collapse
|
5
|
Wan Z, Zhang X, Jia X, Qin Y, Sun N, Xin J, Zeng Y, Jing B, Fang J, Pan K, Zeng D, Bai Y, Wang H, Ma H, Ni X. Lactobacillus johnsonii YH1136 plays a protective role against endogenous pathogenic bacteria induced intestinal dysfunction by reconstructing gut microbiota in mice exposed at high altitude. Front Immunol 2022; 13:1007737. [PMID: 36304467 PMCID: PMC9592553 DOI: 10.3389/fimmu.2022.1007737] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Intestinal microbiota plays an important role in maintaining the microecological balance of the gastrointestinal tract in various animals. Disturbances in the intestinal microbiota may lead to the proliferation of potentially pathogenic bacteria that become the dominant species, leading to intestinal immune disorders, intestinal inflammation, and other intestinal diseases. Numerous studies have been confirmed that high-altitude exposure affects the normal function of the intestine and the composition of the intestinal microbiota. However, it is still necessary to reveal the changes in intestinal microbiota in high-altitude exposure environments, and clarify the relationship between the proliferation of potentially pathogenic bacteria and intestinal injury in this environment. In addition, explored probiotics that may have preventive effects against intestinal diseases. Methods and results C57BL/6 mice were randomly divided into three groups, a high-altitude group (HA), control group (C), and high-altitude probiotic group (HAP). The HA and HAP groups were subjected to hypoxia modeling for 14 days in a low-pressure oxygen chamber with daily gavage of 0.2 mL of normal saline (HA) and Lactobacillus johnsonii YH1136 bacterial fluid (HAP), while the control group was fed normally. L. johnsonii YH1136 was isolated from feces of a healthy Tibetan girl in Baingoin county, the Nagqu region of the Tibet Autonomous Region, at an altitude of 5000 meters. Our observations revealed that gavage of YH1136 was effective in improving the damage to the intestinal barrier caused by high-altitude exposure to hypoxic environments and helped to reduce the likelihood of pathogenic bacteria infection through the intestinal barrier. It also positively regulates the intestinal microbiota to the extent of Lactobacillus being the dominant microbiome and reducing the number of pathogenic bacteria. By analyzing the expression profile of ileal microRNAs and correlation analysis with intestinal microbiota, we found that Staphylococcus and Corynebacterium1 cooperated with miR-196a-1-3p and miR-3060-3p, respectively, to play a regulatory role in the process of high-altitude hypoxia-induced intestinal injury. Conclusion These findings revealed the beneficial effect of L. johnsonii YH1136 in preventing potential endogenous pathogenic bacteria-induced intestinal dysfunction in high-altitude environments. The mechanism may be related to the regulation of intestinal injury from the perspective of the gut microbiota as well as miRNAs.
Collapse
Affiliation(s)
- Zhiqiang Wan
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Xufei Zhang
- Plateau Brain Science Research Center, Tibet University, Lhasa, China
| | - Xianhao Jia
- Plateau Brain Science Research Center, Tibet University, Lhasa, China
| | - Yuhua Qin
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Jinge Xin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Jing Fang
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Yang Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hesong Wang
- Guangzhou Beneco Biotechnology Co. Ltd., Guangzhou, China
| | - Hailin Ma
- Plateau Brain Science Research Center, Tibet University, Lhasa, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
6
|
Quach A, Jayaratne RR, Lee BJ, Ibeawuchi SR, Lim E, Das S, Barrett KE. Diarrheal pathogenesis in Salmonella infection may result from an imbalance in intestinal epithelial differentiation through reduced Notch signaling. J Physiol 2022; 600:1851-1865. [PMID: 35100665 DOI: 10.1113/jp282585] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/20/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Salmonella is a leading foodborne pathogen known to cause high chloride content diarrhea. Salmonella infection of murine enteroid-derived monolayers decreased DRA expression. Salmonella infection resulted in upregulation of the secretory epithelial marker ATOH1, the goblet cell marker Muc2, and the enteroendocrine cell marker ChgA. Downregulation of DRA may result from infection-induced Notch inhibition, as reflected by decreased expression of Notch intracellular domain and Hes1, as well as from decreased HNF1α signaling. The imbalance in intestinal epithelial differentiation favoring secretory over absorptive cell types is a possible mechanism by which Salmonella elicits diarrhea and may be relevant therapeutically. ABSTRACT Infections with non-typhoidal Salmonella spp. represent the most burdensome foodborne illnesses worldwide, yet despite their prevalence, the mechanism through which Salmonella elicits diarrhea is not entirely known. Intestinal ion transporters play important roles in fluid and electrolyte homeostasis in the intestine. We have previously shown that infection with Salmonella caused decreased colonic expression of the chloride/bicarbonate exchanger SLC26A3 (Down-Regulated in Adenoma; DRA) in a mouse model. In this study, we focused on the mechanism of DRA downregulation during Salmonella infection, by using murine epithelial enteroid-derived monolayers (EDM). The decrease in DRA expression caused by infection was recapitulated in EDM and accompanied by increased expression of ATOH1, the goblet cell marker Muc2, and the enteroendocrine cell marker ChgA. This suggested biased epithelial differentiation towards the secretory, rather than absorptive phenotype. In addition, the downstream Notch effector, Notch Intracellular Domain (NICD) and Hes1 were decreased following Salmonella infection. The relevance of Notch signaling was further investigated using a γ-secretase inhibitor, which recapitulated the downregulation in Hes1 and DRA as well as upregulation in ATOH1 and Muc2 seen following infection. Our findings suggest that Salmonella infection may result in a shift from absorptive to secretory cell types through Notch inhibition, which explains why there is a decreased capacity for absorption and ultimately the accumulation of diarrheal fluid. Our work also shows the value of EDM as a model to investigate mechanisms that might be targeted for therapy of diarrhea caused by Salmonella infection. Abstract figure legend Upon infection of the intestinal epithelium with Salmonella, diarrhea may be explained by an imbalance of intestinal epithelial differentiation. Downregulation of cell-fate commitment to the absorptive lineage, as reflected by decreased Hes1 and DRA, was observed. Conversely, upregulation of epithelial differentiation into secretory cell types was observed, as reflected by increased ATOH1, Muc2, and ChgA. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Andrew Quach
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Rashini R Jayaratne
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Beom Jae Lee
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA.,Department of Gastroenterology, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
| | - Stella-Rita Ibeawuchi
- Department of Pathology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Eileen Lim
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Kim E Barrett
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA.,Current affiliation: UC Davis School of Medicine, Education Building, 4610 X Street, Sacramento, CA, 95817, USA
| |
Collapse
|
7
|
Hardesty JE, Warner JB, Song YL, Rouchka EC, Chen CY, Kang JX, McClain CJ, Warner DR, Kirpich IA. Transcriptional signatures of the small intestinal mucosa in response to ethanol in transgenic mice rich in endogenous n3 fatty acids. Sci Rep 2020; 10:19930. [PMID: 33199802 PMCID: PMC7670449 DOI: 10.1038/s41598-020-76959-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022] Open
Abstract
The intestine interacts with many factors, including dietary components and ethanol (EtOH), which can impact intestinal health. Previous studies showed that different types of dietary fats can modulate EtOH-induced changes in the intestine; however, mechanisms underlying these effects are not completely understood. Here, we examined intestinal transcriptional responses to EtOH in WT and transgenic fat-1 mice (which endogenously convert n6 to n3 polyunsaturated fatty acids [PUFAs]) to identify novel genes and pathways involved in EtOH-associated gut pathology and discern the impact of n3 PUFA enrichment. WT and fat-1 mice were chronically fed EtOH, and ileum RNA-seq and bioinformatic analyses were performed. EtOH consumption led to a marked down-regulation of genes encoding digestive and xenobiotic-metabolizing enzymes, and transcription factors involved in developmental processes and tissue regeneration. Compared to WT, fat-1 mice exhibited a markedly plastic transcriptome response to EtOH. Cell death, inflammation, and tuft cell markers were downregulated in fat-1 mice in response to EtOH, while defense responses and PPAR signaling were upregulated. This transcriptional reprogramming may contribute to the beneficial effects of n3 PUFAs on EtOH-induced intestinal pathology. In summary, our study provides a reference dataset of the intestinal mucosa transcriptional responses to chronic EtOH exposure for future hypothesis-driven mechanistic studies.
Collapse
Affiliation(s)
- Josiah E Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, 505 Hancock St., Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jeffrey B Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, 505 Hancock St., Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Ying L Song
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, 505 Hancock St., Louisville, KY, 40202, USA
| | - Eric C Rouchka
- Department of Computer Science and Engineering, Speed School of Engineering, University of Louisville, Louisville, KY, USA
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, 505 Hancock St., Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, USA
- University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA
- Robley Rex Veterans Medical Center, Louisville, KY, USA
| | - Dennis R Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, 505 Hancock St., Louisville, KY, 40202, USA
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, 505 Hancock St., Louisville, KY, 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
- University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, USA.
- University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
8
|
Oliveira LP, Guimarães VHD, Oliveira JR, Guimarães ALS, de Paula AMB, Bader M, Santos RASD, Santos SHS. Genetic deletion of the angiotensin-(1-7) receptor Mas leads to alterations in gut villi length modulating TLR4/PI3K/AKT and produces microbiome dysbiosis. Neuropeptides 2020; 82:102056. [PMID: 32505463 DOI: 10.1016/j.npep.2020.102056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/17/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023]
Abstract
Renin-Angiotensin System (RAS) is an important peptide cascade involved in physiological processes. RAS homeostasis disruption produces several cardiovascular and metabolic disorders, such as arterial hypertension, atherosclerosis, acute myocardial infarct, obesity, diabetes, metabolic syndrome and increases gastrointestinal tract (GIT) cell proliferation. Angiotensin (Ang)-(1-7) peptide is the main RAS counter-regulatory axis effector. It is formed from ACE2 enzyme and acts mainly through Mas receptor (MasR). In this context, the aim of the present study was to evaluate alterations in small intestine morphology and intestinal microbiota composition in MasR knockout C57BL/6 mice. We analyzed glucose tolerance; insulin sensitivity and blood collected for biochemical parameters as well as small intestine tissues samples for immunohistochemistry. mRNA and bacteria gDNA expression evaluation. mRNA expression was evaluated by qRT-PCR for TLR4, PI3K and AKT. The main results showed that Mas-R-knockout mice presented lower body weight. MasR-knockout mice also presented increased fasted blood glucose and total cholesterol with reduced HDL, lower glucose tolerance and impaired insulin sensitivity. Increased intestinal mucosa length, increased intestinal villi, reduced Lieberkühn crypt depth. The increased expression of cell proliferation markers Ki-67 and Cyclin D1 and increased TLR4, PI3K and AKT expressions were observed with augmented Bacteroidetes and decreased amount of Firmicutes. That results suggests that MasR deletion generated changes in intestinal microbiota, possibly due to a lower neutral amino acids absorption followed by a compensatory increase in intestinal villi length associated with disbiosis and LPS overproduction that ultimately lead to proliferation and cell inflammation.
Collapse
Affiliation(s)
- Luis Paulo Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Victor Hugo Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Janaina Ribeiro Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - André Luiz Sena Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Alfredo Maurício Batista de Paula
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Robson Augusto Souza Dos Santos
- Institute of Biological Sciences (ICB), Physiology Department, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences (ICA), Food Engineering, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
9
|
Zheng L, Rui C, Zhang H, Chen J, Jia X, Xiao Y. Sonic hedgehog signaling in epithelial tissue development. Regen Med Res 2019; 7:3. [PMID: 31898580 PMCID: PMC6941452 DOI: 10.1051/rmr/190004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022] Open
Abstract
The Sonic hedgehog (SHH) signaling pathway is essential for embryonic development and tissue regeneration. The dysfunction of SHH pathway is involved in a variety of diseases, including cancer, birth defects, and other diseases. Here we reviewed recent studies on main molecules involved in the SHH signaling pathway, specifically focused on their function in epithelial tissue and appendages development, including epidermis, touch dome, hair, sebaceous gland, mammary gland, tooth, nail, gastric epithelium, and intestinal epithelium. The advance in understanding the SHH signaling pathway will give us more clues to the mechanisms of tissue repair and regeneration, as well as the development of new treatment for diseases related to dysregulation of SHH signaling pathway.
Collapse
Affiliation(s)
- Lu Zheng
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Chen Rui
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Hao Zhang
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Jing Chen
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Xiuzhi Jia
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Ying Xiao
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| |
Collapse
|
10
|
Wang J, Lu H, Wang W, Zheng N, Wang Y, Hu Z, Ji G. Hepatocyte Nuclear Factor 3β Plays a Suppressive Role in Colorectal Cancer Progression. Front Oncol 2019; 9:1096. [PMID: 31696055 PMCID: PMC6817462 DOI: 10.3389/fonc.2019.01096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/04/2019] [Indexed: 11/17/2022] Open
Abstract
Background and Objective: Hepatocyte nuclear factor 3β (HNF3β) is a key transcription factor in the development of the gastrointestinal tract. However, only few studies have examined its' expression, function and potential clinical significance in colorectal cancer tumorigenesis and progression. Methods: HNF3β expression in colorectal cancer tissue samples of 174 patients was assessed by immunohistochemistry. The results were analyzed with respect to patients' clinicopathological characteristics and survival. Following the in vitro cell transfection, MTT, wound healing, and Transwell assays were used to test cell proliferation, migration, and invasion, respectively. Western blot was used to examine IL6, JAK1, and STAT3 protein expression. The potential for tumor formation was evaluated using a mouse xenograft model. Results: HNF3β expression was lower in colon cancer tissue compared to normal tissue and correlated with UICC clinical stage (P = 0.001), depth of invasion (P = 0.004), regional lymph node metastasis (P = 0.007), distant metastasis (P = 0.048), and poor survival (P < 0.001) in patients with colorectal cancer. Furthermore, HNF3β overexpression impeded proliferation, migration and invasion of SW480 cells via JAK-STAT3 signaling in vitro. Moreso, HNF3β overexpression showed a significant growth inhibition of subcutaneous xenograft tumors in vivo. Conclusions: The results show that HNF3β acts as a suppressor of colorectal cancer progression and decreased HNF3 β expression is closely related to the poor prognosis. Thus, HNF3β may be a potential molecular target for inhibition of colorectal cancer cells and development of new anti-tumor therapies.
Collapse
Affiliation(s)
- Juan Wang
- State Key Laboratory of Cancer Biology, Department of Digestive Surgery, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hao Lu
- Department of General Surgery, Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Wei Wang
- State Key Laboratory of Cancer Biology, Department of Pharmaceutical and Pharmacy Administration, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Nanxin Zheng
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yi Wang
- Department of General Surgery, Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhiqian Hu
- Department of General Surgery, Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Gang Ji
- State Key Laboratory of Cancer Biology, Department of Digestive Surgery, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Li CJ, Lin S, Ranilla-García MJ, Baldwin RL. Transcriptomic Profiling of Duodenal Epithelium Reveals Temporally Dynamic Impacts of Direct Duodenal Starch-Infusion During Dry Period of Dairy Cattle. Front Vet Sci 2019; 6:214. [PMID: 31312641 PMCID: PMC6614288 DOI: 10.3389/fvets.2019.00214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 06/14/2019] [Indexed: 12/24/2022] Open
Abstract
Previous research has demonstrated a positive relationship between dietary Metabolisable Energy Intake (MEI) and increased maintenance energy costs associated with the visceral tissues. Limitations in understanding this relationship include a lack of access to samples to assess regulatory control of the putative response gastrointestinal tissues to nutrients. This experiment was conducted with a single nutrient (starch hydrolysate) infused (7 d) directly into the intestine to mimic typical changes in post-ruminal starch delivery in dairy production settings. Duodenal epithelial samples collected via biopsy were evaluated using next-generation sequencing technology (RNA-Seq) to validate the use of this approach for the profiling and comparison of the transcriptome of cattle intestinal epithelial tissues. Samples of intestinal epithelial tissue were collected prior to and during the infusion of starch hydrolysate. Biopsies were collected on day 0 before and day 1, day 3, and day 7 during the infusion. Additionally, samples were collected on day 1 and day 7 after infusion was discontinued (Day 8 and Day14 of the experiment). Evaluation of RNA-seq data revealed dynamic changes in global gene expression during infusion. On day 7 of the infusion, 1490 genes were found to be differentially expressed (DE) compared to the day 0 control samples with FDR p < 0.05, vs. 105 genes on day 1 and 246 genes on Day 3. However, on day 8, after infusion was terminated for 24 h, only 428 genes were identified as differentially expressed compared to day 0 and only 107 genes continued to be identified by Day 14. Thus, the apparent differential expression of these genes is putatively a result of the single nutrient infused. Further, performing function and pathway analysis of the identified DE genes using IPA, we observe changes in digestive system development, and function pathways are among the primary functions of the DE genes, as well as immune response elements. Finally, primary transcription regulators such as PTH, JUN, WNT, and TNFRSF11B were identified as the activated upstream regulators for specific future focus. Using a serial biopsy approach we are able to identify differentially expressed genes from cow duodenal epithelial tissue in response to a short-term perturbation with infused starch hydrolysate.
Collapse
Affiliation(s)
- Cong-Jun Li
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, Beltsville Agricultural Research Center, USDA, Beltsville, MD, United States
| | - Shudai Lin
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, Beltsville Agricultural Research Center, USDA, Beltsville, MD, United States.,Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - María Jose Ranilla-García
- Departamento de Producción Animal, Instituto de Ganadería de Montaña, CSIC-Universidad de León, Campus de Vegazana, León, Spain
| | - Ransom L Baldwin
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, Beltsville Agricultural Research Center, USDA, Beltsville, MD, United States
| |
Collapse
|
12
|
Fair KL, Colquhoun J, Hannan NRF. Intestinal organoids for modelling intestinal development and disease. Philos Trans R Soc Lond B Biol Sci 2018; 373:20170217. [PMID: 29786552 PMCID: PMC5974440 DOI: 10.1098/rstb.2017.0217] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2018] [Indexed: 12/17/2022] Open
Abstract
Gastrointestinal diseases are becoming increasingly prevalent in developed countries. Immortalized cells and animal models have delivered important but limited insight into the mechanisms that initiate and propagate these diseases. Human-specific models of intestinal development and disease are desperately needed that can recapitulate structure and function of the gut in vitro Advances in pluripotent stem cells and primary tissue culture techniques have made it possible to culture intestinal epithelial cells in three dimensions that self-assemble to form 'intestinal organoids'. These organoids allow for new, human-specific models that can be used to gain insight into gastrointestinal disease and potentially deliver new therapies to treat them. Here we review current in vitro models of intestinal development and disease, considering where improvements could be made and potential future applications in the fields of developmental modelling, drug/toxicity testing and therapeutic uses.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Kathryn L Fair
- Division of Cancer and Stem Cells, School of Medicine, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jennifer Colquhoun
- Division of Cancer and Stem Cells, School of Medicine, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nicholas R F Hannan
- Division of Cancer and Stem Cells, School of Medicine, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
- National Institute for Health Research (NIHR) Nottingham Digestive Diseases Biomedical Research Unit, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
13
|
Jung KB, Lee H, Son YS, Lee JH, Cho HS, Lee MO, Oh JH, Lee J, Kim S, Jung CR, Kim J, Son MY. In vitro and in vivo imaging and tracking of intestinal organoids from human induced pluripotent stem cells. FASEB J 2018; 32:111-122. [PMID: 28855280 DOI: 10.1096/fj.201700504r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/14/2017] [Indexed: 12/16/2022]
Abstract
Human intestinal organoids (hIOs) derived from human pluripotent stem cells (hPSCs) have immense potential as a source of intestines. Therefore, an efficient system is needed for visualizing the stage of intestinal differentiation and further identifying hIOs derived from hPSCs. Here, 2 fluorescent biosensors were developed based on human induced pluripotent stem cell (hiPSC) lines that stably expressed fluorescent reporters driven by intestine-specific gene promoters Krüppel-like factor 5 monomeric Cherry (KLF5mCherry) and intestine-specific homeobox enhanced green fluorescence protein (ISXeGFP). Then hIOs were efficiently induced from those transgenic hiPSC lines in which mCherry- or eGFP-expressing cells, which appeared during differentiation, could be identified in intact living cells in real time. Reporter gene expression had no adverse effects on differentiation into hIOs and proliferation. Using our reporter system to screen for hIO differentiation factors, we identified DMH1 as an efficient substitute for Noggin. Transplanted hIOs under the kidney capsule were tracked with fluorescence imaging (FLI) and confirmed histologically. After orthotopic transplantation, the localization of the hIOs in the small intestine could be accurately visualized using FLI. Our study establishes a selective system for monitoring the in vitro differentiation and for tracking the in vivo localization of hIOs and contributes to further improvement of cell-based therapies and preclinical screenings in the intestinal field.-Jung, K. B., Lee, H., Son, Y. S., Lee, J. H., Cho, H.-S., Lee, M.-O., Oh, J.-H., Lee, J., Kim, S., Jung, C.-R., Kim, J., Son, M.-Y. In vitro and in vivo imaging and tracking of intestinal organoids from human induced pluripotent stem cells.
Collapse
Affiliation(s)
- Kwang Bo Jung
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Hana Lee
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Ye Seul Son
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Ji Hye Lee
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Hyun-Soo Cho
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Mi-Ok Lee
- Immunotherapy Covergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jung-Hwa Oh
- Korea Institute of Toxicology, Daejeon, South Korea; and
| | - Jaemin Lee
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Seokho Kim
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Cho-Rok Jung
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea,
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Mi-Young Son
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea,
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
14
|
Lee SH, Rhee M, Kim JW, Yoon KH. Generation of Insulin-Expressing Cells in Mouse Small Intestine by Pdx1, MafA, and BETA2/NeuroD. Diabetes Metab J 2017; 41:405-416. [PMID: 29086539 PMCID: PMC5663680 DOI: 10.4093/dmj.2017.41.5.405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/03/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND To develop surrogate insulin-producing cells for diabetes therapy, adult stem cells have been identified in various tissues and studied for their conversion into β-cells. Pancreatic progenitor cells are derived from the endodermal epithelium and formed in a manner similar to gut progenitor cells. Here, we generated insulin-producing cells from the intestinal epithelial cells that induced many of the specific pancreatic transcription factors using adenoviral vectors carrying three genes: PMB (pancreatic and duodenal homeobox 1 [Pdx1], V-maf musculoaponeurotic fibrosarcoma oncogene homolog A [MafA], and BETA2/NeuroD). METHODS By direct injection into the intestine through the cranial mesenteric artery, adenoviruses (Ad) were successfully delivered to the entire intestine. After virus injection, we could confirm that the small intestine of the mouse was appropriately infected with the Ad-Pdx1 and triple Ad-PMB. RESULTS Four weeks after the injection, insulin mRNA was expressed in the small intestine, and the insulin gene expression was induced in Ad-Pdx1 and Ad-PMB compared to control Ad-green fluorescent protein. In addition, the conversion of intestinal cells into insulin-expressing cells was detected in parts of the crypts and villi located in the small intestine. CONCLUSION These data indicated that PMB facilitate the differentiation of mouse intestinal cells into insulin-expressing cells. In conclusion, the small intestine is an accessible and abundant source of surrogate insulin-producing cells.
Collapse
Affiliation(s)
- So Hyun Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Marie Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Won Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kun Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
15
|
Ducarouge B, Pelissier-Rota M, Powell R, Buisson A, Bonaz B, Jacquier-Sarlin M. Involvement of CRF2 signaling in enterocyte differentiation. World J Gastroenterol 2017; 23:5127-5145. [PMID: 28811708 PMCID: PMC5537180 DOI: 10.3748/wjg.v23.i28.5127] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/06/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the role of corticotropin releasing factor receptor (CRF2) in epithelial permeability and enterocyte cell differentiation.
METHODS For this purpose, we used rat Sprague Dawley and various colon carcinoma cell lines (SW620, HCT8R, HT-29 and Caco-2 cell lines). Expression of CRF2 protein was analyzed by fluorescent immunolabeling in normal rat colon and then by western blot in dissociated colonic epithelial cells and in the lysates of colon carcinoma cell lines or during the early differentiation of HT-29 cells (ten first days). To assess the impact of CRF2 signaling on colonic cell differentiation, HT-29 and Caco-2 cells were exposed to Urocortin 3 recombinant proteins (Ucn3, 100 nmol/L). In some experiments, cells were pre-exposed to the astressin 2b (A2b) a CRF2 antagonist in order to inhibit the action of Ucn3. Intestinal cell differentiation was first analyzed by functional assays: the trans-cellular permeability and the para-cellular permeability were determined by Dextran-FITC intake and measure of the transepithelial electrical resistance respectively. Morphological modifications associated to epithelial dysfunction were analyzed by confocal microscopy after fluorescent labeling of actin (phaloidin-TRITC) and intercellular adhesion proteins such as E-cadherin, p120ctn, occludin and ZO-1. The establishment of mature adherens junctions (AJ) was monitored by following the distribution of AJ proteins in lipid raft fractions, after separation of cell lysates on sucrose gradients. Finally, the mRNA and the protein expression levels of characteristic markers of intestinal epithelial cell (IEC) differentiation such as the transcriptional factor krüppel-like factor 4 (KLF4) or the dipeptidyl peptidase IV (DPPIV) were performed by RT-PCR and western blot respectively. The specific activities of DPPIV and alkaline phosphatase (AP) enzymes were determined by a colorimetric method.
RESULTS CRF2 protein is preferentially expressed in undifferentiated epithelial cells from the crypts of colon and in human colon carcinoma cell lines. Furthermore, CRF2 expression is down regulated according to the kinetic of HT-29 cell differentiation. By performing functional assays, we found that Ucn3-induced CRF2 signaling alters both para- and trans-cellular permeability of differentiated HT-29 and Caco-2 cells. These effects are partly mediated by Ucn3-induced morphological changes associated with the disruption of mature AJ in HT-29 cells and tight junctions (TJ) in Caco-2 cells. Ucn3-mediated activation of CRF2 decreases mRNA and protein expression levels of KLF4 a transcription factor involved in IEC differentiation. This signaling is correlated to a down-regulation of key IEC markers such as DPPIV and AP, at both transcriptional and post-transcriptional levels.
CONCLUSION Our findings suggest that CRF2 signaling could modulate IEC differentiation. These mechanisms could be relevant to the stress induced epithelial alterations found in inflammatory bowel diseases.
Collapse
|
16
|
Reelin protects from colon pathology by maintaining the intestinal barrier integrity and repressing tumorigenic genes. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2126-2134. [PMID: 28572005 DOI: 10.1016/j.bbadis.2017.05.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/15/2017] [Accepted: 05/29/2017] [Indexed: 12/17/2022]
Abstract
We previously reported that reelin, an extracellular matrix protein first known for its key role in neuronal migration, reduces the susceptibility to dextran sulphate sodium (DSS)-colitis. The aim of the current study was to determine whether reelin protects from colorectal cancer and how reelin defends from colon pathology. In the colon of wild-type and of mice lacking reelin (reeler mice) we have analysed the: i) epithelium cell renewal processes, ii) morphology, iii) Sox9, Cdx2, Smad5, Cyclin D1, IL-6 and IFNγ mRNA abundance in DSS-treated and untreated mice, and iv) development of azoxymethane/DSS-induced colorectal cancer, using histological and real time-PCR methodologies. The reeler mutation increases colitis-associated tumorigenesis, with increased tumours number and size. It also impairs the intestinal barrier because it reduces cell proliferation, migration, differentiation and apoptosis; decreases the number and maturation of goblet cells, and expands the intercellular space of the desmosomes. The intestinal barrier impairment might explain the increased susceptibility to colon pathology exhibited by the reeler mice and is at least mediated by the down-regulation of Sox9 and Cdx2. In response to DSS-colitis, the reeler colon increases the mRNA abundance of IL-6, Smad5 and Cyclin D1 and decreases that of IFNγ, conditions that might result in the increased colitis-associated tumorigenesis found in the reeler mice. In conclusion, the results highlight a role for reelin in maintaining intestinal epithelial cell homeostasis and providing resistance against colon pathology.
Collapse
|
17
|
Lechuga S, Naydenov NG, Feygin A, Jimenez AJ, Ivanov AI. A vesicle trafficking protein αSNAP regulates Paneth cell differentiation in vivo. Biochem Biophys Res Commun 2017; 486:951-957. [PMID: 28359759 DOI: 10.1016/j.bbrc.2017.03.135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/24/2017] [Indexed: 12/20/2022]
Abstract
A soluble N-ethylmaleimide-sensitive factor-attachment protein alpha (αSNAP) is a multifunctional scaffolding protein that regulates intracellular vesicle trafficking and signaling. In cultured intestinal epithelial cells, αSNAP has been shown to be essential for cell survival, motility, and adhesion; however, its physiologic functions in the intestinal mucosa remain unknown. In the present study, we used a mouse with a spontaneous hydrocephalus with hop gait (hyh) mutation of αSNAP to examine the roles of this trafficking protein in regulating intestinal epithelial homeostasis in vivo. Homozygous hyh mice demonstrated decreased expression of αSNAP protein in the intestinal epithelium, but did not display gross abnormalities of epithelial architecture in the colon and ileum. Such αSNAP depletion attenuated differentiation of small intestinal epithelial enteroids ex vivo. Furthermore, αSNAP-deficient mutant animals displayed reduced formation of lysozyme granules in small intestinal crypts and decreased expression of lysozyme and defensins in the intestinal mucosa, which is indicative of defects in Paneth cell differentiation. By contrast, development of Goblet cells, enteroendocrine cells, and assembly of enterocyte apical junctions was not altered in hyh mutant mice. Our data revealed a novel role of αSNAP in the intestinal Paneth cell differentiation in vivo.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Nayden G Naydenov
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Alex Feygin
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Antonio J Jimenez
- Department of Cell Biology, Genetics, and Physiology, University of Malaga, Malaga, Spain
| | - Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
18
|
Comincini S, Manai F, Meazza C, Pagani S, Martinelli C, Pasqua N, Pelizzo G, Biggiogera M, Bozzola M. Identification of Autophagy-Related Genes and Their Regulatory miRNAs Associated with Celiac Disease in Children. Int J Mol Sci 2017; 18:ijms18020391. [PMID: 28208686 PMCID: PMC5343926 DOI: 10.3390/ijms18020391] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/27/2017] [Accepted: 02/06/2017] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CD) is a severe genetic autoimmune disorder, affecting about one in 100 people, where the ingestion of gluten leads to damage in the small intestine. Diagnosing CD is quite complex and requires blood tests and intestinal biopsy examinations. Controversy exists regarding making the diagnosis without biopsy, due to the large spectrum of manifesting symptoms; furthermore, small-intestinal gastroscopy examinations have a relatively complex management in the pediatric population. To identify novel molecular markers useful to increase the sensitivity and specificity in the diagnosis of pediatric CD patients, the expression levels of two key autophagy executor genes (ATG7 and BECN1) and their regulatory validated miRNAs (miR-17 and miR-30a, respectively) were analyzed by relative quantitative real-time-PCR on a cohort of confirmed CD patients compared to age-related controls. Among the investigated targets, the non-parametric Mann–Whitney U test and ROC analysis indicated the highest significant association of BECN1 with CD status in the blood, while in intestinal biopsies, all of the investigated sequences were positively associated with CD diagnosis. Nomogram-based analysis showed nearly opposite expression trends in blood compared to intestine tissue, while hierarchical clustering dendrograms enabled identifying CD and control subgroups based on specific genes and miRNA expression signatures. Next, using an established in vitro approach, through digested gliadin administration in Caco-2 cells, we also highlighted that the modulation of miR-17 endogenous levels using enriched exosomes increased the intracellular autophagosome content, thereby altering the autophagic status. Altogether, these results highlighted novel molecular markers that might be useful to increase the accuracy in CD diagnosis and in molecular-based stratification of the patients, further reinforcing the functional involvement of the regulation of the autophagy process within a digestive and autoimmune-related disorder as CD.
Collapse
Affiliation(s)
- Sergio Comincini
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Federico Manai
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Cristina Meazza
- Pediatrics and Adolescentology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Fondazione IRCCS San Matteo, 27100 Pavia, Italy.
| | - Sara Pagani
- Pediatrics and Adolescentology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Fondazione IRCCS San Matteo, 27100 Pavia, Italy.
| | - Carolina Martinelli
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Noemi Pasqua
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Fondazione IRCCS San Matteo, 27100 Pavia, Italy.
| | - Gloria Pelizzo
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Fondazione IRCCS San Matteo, 27100 Pavia, Italy.
| | - Marco Biggiogera
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Mauro Bozzola
- Pediatrics and Adolescentology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Fondazione IRCCS San Matteo, 27100 Pavia, Italy.
| |
Collapse
|
19
|
Sala P, Belarmino G, Machado NM, Cardinelli CS, Al Assal K, Silva MM, Fonseca DC, Ishida RK, Santo MA, de Moura EGH, Sakai P, Guarda IFMS, da Silva IDCG, Rodrigues AS, Pereira CADB, Heymsfield S, Doré J, Torrinhas RSMDM, Giannella-Neto D, Waitzberg DL. The SURMetaGIT study: Design and rationale for a prospective pan-omics examination of the gastrointestinal response to Roux-en-Y gastric bypass surgery. J Int Med Res 2016; 44:1359-1375. [PMID: 27834300 PMCID: PMC5536762 DOI: 10.1177/0300060516667862] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Objective To describe the protocol of the SURgically induced Metabolic effects on the Human GastroIntestinal Tract (SURMetaGIT) study, a clinical pan-omics study exploring the gastrointestinal tract as a central organ driving remission of type 2 diabetes mellitus (T2DM) after Roux-en-Y gastric bypass (RYGB). The main points considered in the study’s design and challenges faced in its application are detailed. Methods This observational, longitudinal, prospective study involved collection of gastrointestinal biopsy specimens, faeces, urine, and blood from 25 obese women with T2DM who were candidates for RYGB (20 patients for omics assessment and 5 for omics validation). These collections were performed preoperatively and 3 and 24 months postoperatively. Gastrointestinal transcriptomics; faecal metagenomics and metabolomics; plasma proteomics, lipidomics, and metabolomics; and biochemical, nutritional, and metabolic data were assessed to identify their short- and long-term correlations with T2DM remission. Results Data were collected from 20 patients before and 3 months after RYGB. These patients have nearly completed the 2-year follow-up assessments. The five additional patients are currently being selected for omics data validation. Conclusion The multi-integrated pan-omics approach of the SURMetaGIT study enables integrated analysis of data that will contribute to the understanding of molecular mechanisms involved in T2DM remission after RYGB.
Collapse
Affiliation(s)
- Priscila Sala
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
- Priscila Sala - Av. Dr. Arnaldo, 455, 2° andar, sala 2208 – Cerqueira César - CEP: 01246-903, São Paulo – SP, Brazil.
| | - Giliane Belarmino
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Natasha Mendonça Machado
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Camila Siqueira Cardinelli
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Karina Al Assal
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Mariane Marques Silva
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Danielle Cristina Fonseca
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Robson Kiyoshi Ishida
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | - Marco Aurélio Santo
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | | | - Paulo Sakai
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| | | | | | | | | | - Steven Heymsfield
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, LA, USA
| | - Joel Doré
- Institut National de la Recherche Agronomique INRA, France
| | | | | | - Dan Linetzky Waitzberg
- Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, FMUSP – University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Kodama N, Iwao T, Kabeya T, Horikawa T, Niwa T, Kondo Y, Nakamura K, Matsunaga T. Inhibition of mitogen-activated protein kinase kinase, DNA methyltransferase, and transforming growth factor-β promotes differentiation of human induced pluripotent stem cells into enterocytes. Drug Metab Pharmacokinet 2016; 31:193-200. [DOI: 10.1016/j.dmpk.2016.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 01/21/2016] [Accepted: 02/09/2016] [Indexed: 12/13/2022]
|
21
|
Plaul SE, Pastor R, Díaz AO, Barbeito CG. Immunohistochemical and ultrastructural evidence of functional organization along theCorydoras paleatusintestine. Microsc Res Tech 2016; 79:140-8. [DOI: 10.1002/jemt.22614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/02/2015] [Accepted: 12/04/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Silvia E. Plaul
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada, Facultad de Ciencias Veterinarias, UNLP. La Plata; Buenos Aires Argentina
| | - Raquel Pastor
- Departamento de Ciencias Morfológicas; Facultad de Ciencias Veterinarias, UNL. Esperanza; Santa Fe Argentina
| | - Alcira O. Díaz
- Laboratorio de Histología e Histoquímica, Departamento de Biología, Instituto de Investigaciones Marinas y Costeras (IIMyC), Facultad de Ciencias Exactas y Naturales, CONICET, UNdMP. Mar del Plata; Buenos Aires Argentina
| | - Claudio G. Barbeito
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada, Facultad de Ciencias Veterinarias, UNLP. La Plata; Buenos Aires Argentina
| |
Collapse
|
22
|
Bousquet J, Anto JM, Wickman M, Keil T, Valenta R, Haahtela T, Lodrup Carlsen K, van Hage M, Akdis C, Bachert C, Akdis M, Auffray C, Annesi-Maesano I, Bindslev-Jensen C, Cambon-Thomsen A, Carlsen KH, Chatzi L, Forastiere F, Garcia-Aymerich J, Gehrig U, Guerra S, Heinrich J, Koppelman GH, Kowalski ML, Lambrecht B, Lupinek C, Maier D, Melén E, Momas I, Palkonen S, Pinart M, Postma D, Siroux V, Smit HA, Sunyer J, Wright J, Zuberbier T, Arshad SH, Nadif R, Thijs C, Andersson N, Asarnoj A, Ballardini N, Ballereau S, Bedbrook A, Benet M, Bergstrom A, Brunekreef B, Burte E, Calderon M, De Carlo G, Demoly P, Eller E, Fantini MP, Hammad H, Hohman C, Just J, Kerkhof M, Kogevinas M, Kull I, Lau S, Lemonnier N, Mommers M, Nawijn M, Neubauer A, Oddie S, Pellet J, Pin I, Porta D, Saes Y, Skrindo I, Tischer CG, Torrent M, von Hertzen L. Are allergic multimorbidities and IgE polysensitization associated with the persistence or re-occurrence of foetal type 2 signalling? The MeDALL hypothesis. Allergy 2015; 70:1062-78. [PMID: 25913421 DOI: 10.1111/all.12637] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2015] [Indexed: 12/22/2022]
Abstract
Allergic diseases [asthma, rhinitis and atopic dermatitis (AD)] are complex. They are associated with allergen-specific IgE and nonallergic mechanisms that may coexist in the same patient. In addition, these diseases tend to cluster and patients present concomitant or consecutive diseases (multimorbidity). IgE sensitization should be considered as a quantitative trait. Important clinical and immunological differences exist between mono- and polysensitized subjects. Multimorbidities of allergic diseases share common causal mechanisms that are only partly IgE-mediated. Persistence of allergic diseases over time is associated with multimorbidity and/or IgE polysensitization. The importance of the family history of allergy may decrease with age. This review puts forward the hypothesis that allergic multimorbidities and IgE polysensitization are associated and related to the persistence or re-occurrence of foetal type 2 signalling. Asthma, rhinitis and AD are manifestations of a common systemic immune imbalance (mesodermal origin) with specific patterns of remodelling (ectodermal or endodermal origin). This study proposes a new classification of IgE-mediated allergic diseases that allows the definition of novel phenotypes to (i) better understand genetic and epigenetic mechanisms, (ii) better stratify allergic preschool children for prognosis and (iii) propose novel strategies of treatment and prevention.
Collapse
Affiliation(s)
- J. Bousquet
- University Hospital; Montpellier France
- MACVIA-LR; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon; European Innovation Partnership on Active and Healthy Ageing Reference Site; Paris France
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - J. M. Anto
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - M. Wickman
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - T. Keil
- Institute of Social Medicine, Epidemiology and Health Economics; Charité - Universitätsmedizin Berlin; Berlin Germany
- Institute for Clinical Epidemiology and Biometry; University of Wuerzburg; Wuerzburg Germany
| | - R. Valenta
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - T. Haahtela
- Skin and Allergy Hospital; Helsinki University Hospital; Helsinki Finland
| | - K. Lodrup Carlsen
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - M. van Hage
- Clinical Immunology and Allergy Unit; Department of Medicine Solna; Karolinska Institutet and University Hospital; Stockholm Sweden
| | - C. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
| | - C. Bachert
- ENT Department; Ghent University Hospital; Gent Belgium
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
| | - C. Auffray
- European Institute for Systems Biology and Medicine; Lyon France
| | - I. Annesi-Maesano
- EPAR U707 INSERM; Paris France
- EPAR UMR-S UPMC; Paris VI; Paris France
| | - C. Bindslev-Jensen
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | - A. Cambon-Thomsen
- UMR Inserm U1027; Université de Toulouse III Paul Sabatier; Toulouse France
| | - K. H. Carlsen
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- University of Oslo; Oslo Norway
| | - L. Chatzi
- Department of Social Medicine; Faculty of Medicine; University of Crete; Heraklion Crete Greece
| | - F. Forastiere
- Department of Epidemiology; Regional Health Service Lazio Region; Rome Italy
| | - J. Garcia-Aymerich
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - U. Gehrig
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - S. Guerra
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - J. Heinrich
- Institute of Epidemiology; German Research Centre for Environmental Health; Helmholtz Zentrum München; Neuherberg Germany
| | - G. H. Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | - M. L. Kowalski
- Department of Immunology, Rheumatology and Allergy; Medical University of Lodz; Lodz Poland
| | - B. Lambrecht
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - C. Lupinek
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | | | - E. Melén
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - I. Momas
- Department of Public Health and Biostatistics, EA 4064; Paris Descartes University; Paris France
- Paris Municipal Department of Social Action, Childhood, and Health; Paris France
| | - S. Palkonen
- EFA European Federation of Allergy and Airways Diseases Patients' Associations; Brussels Belgium
| | - M. Pinart
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - D. Postma
- Department of Respiratory Medicine; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | | | - H. A. Smit
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - J. Sunyer
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - J. Wright
- Bradford Institute for Health Research; Bradford Royal Infirmary; Bradford UK
| | - T. Zuberbier
- Allergy-Centre-Charité at the Department of Dermatology; Charité - Universitätsmedizin Berlin; Berlin Germany
- Secretary General of the Global Allergy and Asthma European Network (GA2LEN); Berlin Germany
| | - S. H. Arshad
- David Hide Asthma and Allergy Research Centre; Isle of Wight UK
| | - R. Nadif
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - C. Thijs
- Department of Epidemiology; CAPHRI School of Public Health and Primary Care; Maastricht University; Maastricht the Netherlands
| | - N. Andersson
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - A. Asarnoj
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - N. Ballardini
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - S. Ballereau
- European Institute for Systems Biology and Medicine; Lyon France
| | - A. Bedbrook
- MACVIA-LR; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon; European Innovation Partnership on Active and Healthy Ageing Reference Site; Paris France
| | - M. Benet
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - A. Bergstrom
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - B. Brunekreef
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - E. Burte
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - M. Calderon
- National Heart and Lung Institute; Imperial College London; Royal Brompton Hospital NHS; London UK
| | - G. De Carlo
- EFA European Federation of Allergy and Airways Diseases Patients' Associations; Brussels Belgium
| | - P. Demoly
- Department of Respiratory Diseases; Montpellier University Hospital; Montpellier France
| | - E. Eller
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | - M. P. Fantini
- Department of Medicine and Public Health; Alma Mater Studiorum - University of Bologna; Bologna Italy
| | - H. Hammad
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - C. Hohman
- Institute of Social Medicine, Epidemiology and Health Economics; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - J. Just
- Allergology Department; Centre de l'Asthme et des Allergies; Hôpital d'Enfants Armand-Trousseau (APHP); Paris France
- Institut Pierre Louis d'Epidémiologie et de Santé Publique; Equipe EPAR; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136; Paris France
| | - M. Kerkhof
- Department of Respiratory Medicine; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | - M. Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - I. Kull
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - S. Lau
- Department for Pediatric Pneumology and Immunology; Charité Medical University; Berlin Germany
| | - N. Lemonnier
- European Institute for Systems Biology and Medicine; Lyon France
| | - M. Mommers
- Department of Epidemiology; CAPHRI School of Public Health and Primary Care; Maastricht University; Maastricht the Netherlands
| | - M. Nawijn
- Department of Pediatric Pulmonology and Pediatric Allergology; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | | | - S. Oddie
- Bradford Institute for Health Research; Bradford Royal Infirmary; Bradford UK
| | - J. Pellet
- European Institute for Systems Biology and Medicine; Lyon France
| | - I. Pin
- Département de pédiatrie; CHU de Grenoble; Grenoble Cedex 9 France
| | - D. Porta
- Department of Epidemiology; Regional Health Service Lazio Region; Rome Italy
| | - Y. Saes
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - I. Skrindo
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - C. G. Tischer
- Institute of Epidemiology; German Research Centre for Environmental Health; Helmholtz Zentrum München; Neuherberg Germany
| | - M. Torrent
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Area de Salut de Menorca, ib-salut; Illes Balears Spain
| | - L. von Hertzen
- Skin and Allergy Hospital; Helsinki University Hospital; Helsinki Finland
| |
Collapse
|
23
|
Abstract
Epithelial cells are tightly coupled together through specialized intercellular junctions, including adherens junctions, desmosomes, tight junctions, and gap junctions. A growing body of evidence suggests epithelial cells also directly exchange information at cell-cell contacts via the Eph family of receptor tyrosine kinases and their membrane-associated ephrin ligands. Ligand-dependent and -independent signaling via Eph receptors as well as reverse signaling through ephrins impact epithelial tissue homeostasis by organizing stem cell compartments and regulating cell proliferation, migration, adhesion, differentiation, and survival. This review focuses on breast, gut, and skin epithelia as representative examples for how Eph receptors and ephrins modulate diverse epithelial cell responses in a context-dependent manner. Abnormal Eph receptor and ephrin signaling is implicated in a variety of epithelial diseases raising the intriguing possibility that this cell-cell communication pathway can be therapeutically harnessed to normalize epithelial function in pathological settings like cancer or chronic inflammation.
Collapse
Key Words
- ADAM, a disintegrin and metalloprotease
- Apc, adenomatous polyposis coli
- Breast
- ER, estrogen receptor
- Eph receptor
- Eph, erythropoietin-producing hepatocellular
- Erk, extracellular signal-regulated kinase
- GEF, guanine nucleotide exchange factor
- GPI, glycosylphosphatidylinositol
- HER2, human epidermal growth factor receptor 2
- HGF, hepatocyte growth factor
- IBD, inflammatory bowel disease
- KLF, Krüppel-like factor
- MAPK, mitogen-activated protein kinase
- MMTV-LTR, mouse mammary tumor virus-long terminal repeat
- MT1-MMP, membrane-type 1 matrix metalloproteinase
- PDZ, postsynaptic density protein 95, discs large 1, and zonula occludens-1
- PTP, protein tyrosine phosphatase
- RTK, receptor tyrosine kinase
- SH2, Src homology 2
- SHIP2, SH2 inositol phosphatase 2
- SLAP, Src-like adaptor protein
- TCF, T-cell specific transcription factor
- TEB, terminal end bud
- TNFα, tumor necrosis factor α.
- cell-cell
- ephrin
- epithelial
- intestine
- receptor tyrosine kinase
- skin
- stem cell
Collapse
|
24
|
Walsh MF, Hermann R, Lee JH, Chaturvedi L, Basson MD. Schlafen 3 Mediates the Differentiating Effects of Cdx2 in Rat IEC-Cdx2L1 Enterocytes. J INVEST SURG 2015; 28:202-207. [PMID: 26268420 PMCID: PMC4771065 DOI: 10.3109/08941939.2015.1005780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
AIM Mature, differentiated enterocytes are essential for normal gut function and critical to recovery from pathological conditions. Little is known about the factors that regulate intestinal epithelial cell differentiation in the adult intestine. The transcription factor, Cdx2, involved in enterocytic differentiation, remains expressed in the adult. Since we have implicated Slfn3 in differentiation in vivo and in vitro, we examined whether it also mediated differentiation in the IEC-Cdx2-L1 cell model of differentiation. MATERIALS AND METHODS IEC-Cdx2-L1 cells, permanently transfected with Cdx2 under the control of isopropyl-β-D-thiogalactoside (IPTG), were stimulated to differentiate by 16-day exposure to IPTG. Transcript levels of Cdx2, Slfn 3, and villin were determined by quantitative reverse transcriptase-polymerase chain reaction of mRNA isolated from IPTG-treated and control cells. Slfn3 expression was lowered with specific siRNA to investigate the role of Slfn3 in Cdx2-driven villin expression in IPTG-differentiated cells. RESULTS Slfn3 and villin expression were significantly greater in IPTG-treated cells. Slfn3 siRNA lowered Slfn3 expression and abolished the IPTG-induced rise in villin expression (p < .05 by ANOVA); Cdx2 expression was unaffected by Slfn3 siRNA. DISCUSSION The data indicate that the presence of Slfn3 is required for Cdx2 to induce villin expression, and thus differentiation. However, Slfn3 must also promote differentiation of Cdx2 independently since IEC-6 cells that do not normally express Cdx2 can be differentiated by a variety of Slfn3-dependent mechanisms.
Collapse
Affiliation(s)
- Mary F Walsh
- Department of Surgery, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | | | | | | | | |
Collapse
|
25
|
The transcription factor FOXA2 suppresses gastric tumorigenesis in vitro and in vivo. Dig Dis Sci 2015; 60:109-17. [PMID: 25129104 DOI: 10.1007/s10620-014-3290-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/11/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND AIMS The transcription factor forkhead box A2 (FOXA2) plays a central role in the development of endoderm-derived organs. It has been reported that FOXA2 acts as a suppressor in many kinds of tumor. However, little is known about the role of FOXA2 in gastric cancer. METHODS The expression of FOXA2 in gastric cancer tissue samples from 89 patients was assessed by immunohistochemistry, and the clinicopathological characteristics of the samples were analyzed. The human gastric cancer cell line, BGC-823, was used to investigate the effects of FOXA2 in gastric cancer in vitro and in vivo and the potential mechanism involved was explored. RESULTS FOXA2 expression in human gastric cancer cell lines and human gastric cancer tissues was lower compared with the normal gastric epithelium cell line GES1 and normal adult gastric tissues, respectively. Patients with high FOXA2 expression level had longer 5-year overall survival than those with low FOXA2 expression level. FOXA2 markedly inhibited growth of BGC-823 cells accompanied with the cell cycle arrest and apoptosis. Infection of BGC-823 cells by FOXA2 lentivirus resulted in reduced cell tumorigenesis in vitro and in vivo. Moreover, expression of Mucin 5AC was up-regulated along with increased expression of exogenous FOXA2 in BGC-823 cells; in contrast, dedifferentiation markers, BMI, CD54 and CD24, were down-regulated. CONCLUSIONS These results suggest that FOXA2 induces the differentiation of gastric cancer and highlight FOXA2 as a novel therapeutic target and prognostic marker for human gastric cancer.
Collapse
|
26
|
Welch MG, Margolis KG, Li Z, Gershon MD. Oxytocin regulates gastrointestinal motility, inflammation, macromolecular permeability, and mucosal maintenance in mice. Am J Physiol Gastrointest Liver Physiol 2014; 307:G848-62. [PMID: 25147234 PMCID: PMC4200316 DOI: 10.1152/ajpgi.00176.2014] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/18/2014] [Indexed: 01/31/2023]
Abstract
Enteric neurons express oxytocin (OT); moreover, enteric neurons and enterocytes express developmentally regulated OT receptors (OTRs). Although OT (with secretin) opposes intestinal inflammation, physiological roles played by enteric OT/OTR signaling have not previously been determined. We tested hypotheses that OT/OTR signaling contributes to enteric nervous system (ENS)-related gastrointestinal (GI) physiology. GI functions and OT effects were compared in OTR-knockout (OTRKO) and wild-type (WT) mice. Stool mass and water content were greater in OTRKO mice than in WT. GI transit time in OTRKO animals was faster than in WT; OT inhibited in vitro generation of ENS-dependent colonic migrating motor complexes in WT but not in OTRKO mice. Myenteric neurons were hyperplastic in OTRKO animals, and mucosal exposure to cholera toxin (CTX) in vitro activated Fos in more myenteric neurons in OTRKO than WT than in WT mice; OT inhibited the CTX response in WT but not in OTRKO mice. Villi and crypts were shorter in OTRKO than in WT mice, and transit-amplifying cell proliferation in OTRKO crypts was deficient. Macromolecular intestinal permeability in OTRKO was greater than WT mice, and experimental colitis was more severe in OTRKO mice; moreover, OT protected WT animals from colitis. Observations suggest that OT/OTR signaling acts as a brake on intestinal motility, decreases mucosal activation of enteric neurons, and promotes enteric neuronal development and/or survival. It also regulates proliferation of crypt cells and mucosal permeability; moreover OT/OTR signaling is protective against inflammation. Oxytocinergic signaling thus appears to play an important role in multiple GI functions that are subject to neuronal regulation.
Collapse
Affiliation(s)
- Martha G Welch
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Kara G Margolis
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Zhishan Li
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Michael D Gershon
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| |
Collapse
|
27
|
Tümer E, Bröer A, Balkrishna S, Jülich T, Bröer S. Enterocyte-specific regulation of the apical nutrient transporter SLC6A19 (B(0)AT1) by transcriptional and epigenetic networks. J Biol Chem 2013; 288:33813-33823. [PMID: 24121511 DOI: 10.1074/jbc.m113.482760] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Enterocytes are specialized to absorb nutrients from the lumen of the small intestine by expressing a select set of genes to maximize the uptake of nutrients. They develop from stem cells in the crypt and differentiate into mature enterocytes while moving along the crypt-villus axis. Using the Slc6a19 gene as an example, encoding the neutral amino acid transporter B(0)AT1, we studied regulation of the gene by transcription factors and epigenetic factors in the intestine. To investigate this question, we used a fractionation method to separate mature enterocytes from crypt cells and analyzed gene expression. Transcription factors HNF1a and HNF4a activate transcription of the Slc6a19 gene in villus enterocytes, whereas high levels of SOX9 repress expression in the crypts. CpG dinucleotides in the proximal promoter were highly methylated in the crypt and fully de-methylated in the villus. Furthermore, histone modification H3K27Ac, indicating an active promoter, was prevalent in villus cells but barely detectable in crypt cells. The results suggest that Slc6a19 expression in the intestine is regulated at three different levels involving promoter methylation, histone modification, and opposing transcription factors.
Collapse
Affiliation(s)
- Emrah Tümer
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 0200, Australia
| | - Angelika Bröer
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 0200, Australia
| | - Sarojini Balkrishna
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 0200, Australia
| | - Torsten Jülich
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 0200, Australia
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 0200, Australia.
| |
Collapse
|
28
|
Mohan M, Kaushal D, Aye PP, Alvarez X, Veazey RS, Lackner AA. Focused examination of the intestinal epithelium reveals transcriptional signatures consistent with disturbances in enterocyte maturation and differentiation during the course of SIV infection. PLoS One 2013; 8:e60122. [PMID: 23593167 PMCID: PMC3621888 DOI: 10.1371/journal.pone.0060122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 02/21/2013] [Indexed: 12/29/2022] Open
Abstract
The Gastrointestinal (GI) tract plays a pivotal role in AIDS pathogenesis as it is the primary site for viral transmission, replication and CD4+ T cell destruction. Accordingly, GI disease (enteropathy) has become a well-known complication and a driver of AIDS progression. To better understand the molecular mechanisms underlying GI disease we analyzed global gene expression profiles sequentially in the intestinal epithelium of the same animals before SIV infection and at 21 and 90 days post infection (DPI). More importantly we obtained sequential excisional intestinal biopsies and examined distinct mucosal components (epithelium. intraepithelial lymphocytes, lamina propria lymphocytes, fibrovascular stroma) separately. Here we report data pertaining to the epithelium. Overall genes associated with epithelial cell renewal/proliferation/differentiation, permeability and adhesion were significantly down regulated (<1.5–7 fold) at 21 and 90DPI. Genes regulating focal adhesions (n = 6), gap junctions (n = 3), ErbB (n = 3) and Wnt signaling (n = 4) were markedly down at 21DPI and the number of genes in each of these groups that were down regulated doubled between 21 and 90DPI. Notable genes included FAK, ITGA6, PDGF, TGFβ3, Ezrin, FZD6, WNT10A, and TCF7L2. In addition, at 90DPI genes regulating ECM-receptor interactions (laminins and ITGB1), epithelial cell gene expression (PDX1, KLF6), polarity/tight junction formation (PARD3B&6B) and histone demethylase (JMJD3) were also down regulated. In contrast, expression of NOTCH3, notch target genes (HES4, HES7) and EZH2 (histone methyltransferase) were significantly increased at 90DPI. The altered expression of genes linked to Wnt signaling together with decreased expression of PDX1, PARD3B, PARD6B and SDK1 suggests marked perturbations in intestinal epithelial function and homeostasis leading to breakdown of the mucosal barrier. More importantly, the divergent expression patterns of EZH2 and JMJD3 suggests that an epigenetic mechanism involving histone modifications may contribute to the massive decrease in gene expression at 90DPI leading to defects in enterocyte maturation and differentiation.
Collapse
Affiliation(s)
- Mahesh Mohan
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Deepak Kaushal
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Pyone P. Aye
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Xavier Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Andrew A. Lackner
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
29
|
In Vivo Characterization of a Novel γ-Secretase Inhibitor SCH 697466 in Rodents and Investigation of Strategies for Managing Notch-Related Side Effects. Int J Alzheimers Dis 2013; 2013:823528. [PMID: 23573456 PMCID: PMC3612465 DOI: 10.1155/2013/823528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/27/2012] [Indexed: 11/17/2022] Open
Abstract
Substantial evidence implicates β-amyloid (Aβ) peptides in the etiology of Alzheimer's disease (AD). Aβ is produced by the proteolytic cleavage of the amyloid precursor protein by β- and γ-secretase suggesting that γ-secretase inhibition may provide therapeutic benefit for AD. Although many γ-secretase inhibitors have been shown to be potent at lowering Aβ, some have also been shown to have side effects following repeated administration. All of these side effects can be attributed to altered Notch signaling, another γ-secretase substrate. Here we describe the in vivo characterization of the novel γ-secretase inhibitor SCH 697466 in rodents. Although SCH 697466 was effective at lowering Aβ, Notch-related side effects in the intestine and thymus were observed following subchronic administration at doses that provided sustained and complete lowering of Aβ. However, additional studies revealed that both partial but sustained lowering of Aβand complete but less sustained lowering of Aβ were successful approaches for managing Notch-related side effects. Further, changes in several Notch-related biomarkers paralleled the side effect observations. Taken together, these studies demonstrated that, by carefully varying the extent and duration of Aβ lowering by γ-secretase inhibitors, it is possible to obtain robust and sustained lowering of Aβ without evidence of Notch-related side effects.
Collapse
|
30
|
Cheng ASL, Li MS, Kang W, Cheng VY, Chou JL, Lau SS, Go MY, Lee CC, Ling TK, Ng EK, Yu J, Huang TH, To KF, Chan MW, Sung JJY, Chan FKL. Helicobacter pylori causes epigenetic dysregulation of FOXD3 to promote gastric carcinogenesis. Gastroenterology 2013; 144:122-133.e9. [PMID: 23058321 DOI: 10.1053/j.gastro.2012.10.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 08/25/2012] [Accepted: 10/03/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Deregulation of forkhead box (Fox) proteins, an evolutionarily conserved family of transcriptional regulators, leads to tumorigenesis. Little is known about their regulation or functions in the pathogenesis of gastric cancer. Promoter hypermethylation occurs during Helicobacter pylori-induced gastritis. We investigated whether the deregulated genes contribute to gastric tumorigenesis. METHODS We used integrative genome-wide scans to identify concomitant hypermethylated genes in mice infected with H pylori and human gastric cancer samples. We also analyzed epigenetic gene silencing in gastric tissues from patients with H pylori infection and gastritis, intestinal metaplasia, gastric tumors, or without disease (controls). Target genes were identified by chromatin immunoprecipitation microarrays and expression and luciferase reporter analyses. RESULTS Methylation profile analyses identified the promoter of FOXD3 as the only genomic region with increased methylation in mice and humans during progression of H pylori-associated gastric tumors. FOXD3 methylation also correlated with shorter survival times of patients with gastric cancer. Genome demethylation reactivated FOXD3 expression in gastric cancer cell lines. Transgenic overexpression of FOXD3 significantly inhibited gastric cancer cell proliferation and invasion, and reduced growth of xenograft tumors in mice, at least partially, by promoting tumor cell apoptosis. FOXD3 bound directly to the promoters of, and activated transcription of, genes encoding the cell death regulators CYFIP2 and RARB. Levels of FOXD3, CYFIP2, and RARB messenger RNAs were reduced in human gastric tumor samples, compared with control tissues. CONCLUSIONS FOXD3-mediated transcriptional control of tumor suppressors is deregulated by H pylori infection-induced hypermethylation; this could perturb the balance between cell death and survival. These findings identify a pathway by which epigenetic changes affect gastric tumor suppression.
Collapse
Affiliation(s)
- Alfred S L Cheng
- Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Shaw D, Gohil K, Basson MD. Intestinal mucosal atrophy and adaptation. World J Gastroenterol 2012; 18:6357-6375. [PMID: 23197881 PMCID: PMC3508630 DOI: 10.3748/wjg.v18.i44.6357] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 11/06/2012] [Accepted: 11/14/2012] [Indexed: 02/06/2023] Open
Abstract
Mucosal adaptation is an essential process in gut homeostasis. The intestinal mucosa adapts to a range of pathological conditions including starvation, short-gut syndrome, obesity, and bariatric surgery. Broadly, these adaptive functions can be grouped into proliferation and differentiation. These are influenced by diverse interactions with hormonal, immune, dietary, nervous, and mechanical stimuli. It seems likely that clinical outcomes can be improved by manipulating the physiology of adaptation. This review will summarize current understanding of the basic science surrounding adaptation, delineate the wide range of potential targets for therapeutic intervention, and discuss how these might be incorporated into an overall treatment plan. Deeper insight into the physiologic basis of adaptation will identify further targets for intervention to improve clinical outcomes.
Collapse
|
32
|
Yang Y, Tarapore RS, Jarmel MH, Tetreault MP, Katz JP. p53 mutation alters the effect of the esophageal tumor suppressor KLF5 on keratinocyte proliferation. Cell Cycle 2012; 11:4033-9. [PMID: 22990386 DOI: 10.4161/cc.22265] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Krüppel-like factor 5 (KLF5) is a key transcriptional regulator that is typically pro-proliferative in non-transformed epithelial cells but inhibits proliferation in transformed epithelial cells. However, the underlying mechanisms for this context-dependent function are not known. KLF5 is epigenetically silenced and exhibits a tumor suppressive function in esophageal squamous cell cancer (ESCC). Since p53 mutation is the most common genetic alteration in ESCC, as in other human epithelial cancers, we hypothesized that the context-dependent functions of KLF5 in cell proliferation were dependent on p53 status. In fact, in non-transformed human primary esophageal keratinocytes, when p53 was wild-type, KLF5 was pro-proliferative; however, KLF5 became anti-proliferative when p53 was mutated. KLF5 loss in human primary keratinocytes harboring p53 mutation accelerated the cell cycle and decreased expression of p21Waf1/Cip1; similar effects were also seen in ESCC cells with established p53 mutations. Further, p21Waf1/Cip1 was directly and differentially bound and regulated by KLF5 in the presence or absence of mutant p53, and suppression of p21Waf1/Cip1 reversed the antiproliferative effects of KLF5 in the presence of p53 mutation. Thus, KLF5 is a critical brake on an aberrant cell cycle, with important tumor suppressive functions in esophageal squamous cell and potentially other epithelial cancers.
Collapse
Affiliation(s)
- Yizeng Yang
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
33
|
Sirakov M, Skah S, Nadjar J, Plateroti M. Thyroid hormone's action on progenitor/stem cell biology: new challenge for a classic hormone? Biochim Biophys Acta Gen Subj 2012; 1830:3917-27. [PMID: 22890105 DOI: 10.1016/j.bbagen.2012.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 07/01/2012] [Accepted: 07/29/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Thyroid hormones are involved in developmental and homeostatic processes in several tissues. Their action results in different outcomes depending on the developmental stage, tissue and/or cellular context. Interestingly, their pleiotropic roles are conserved across vertebrates. It is largely documented that thyroid hormones act via nuclear receptors, the TRs, which are transcription factors and whose activity can be modulated by the local availability of the hormone T3. In the "classical view", the T3-induced physiological response depends on the expression of specific TR isoforms and the iodothyronine deiodinase selenoenzymes that control the local level of T3, thus TR activity. SCOPE OF THE REVIEW Recent data have clearly established that the functionality of TRs is coordinated and integrated with other signaling pathways, specifically at the level of stem/progenitor cell populations. Here, we summarize these data and propose a new and intriguing role for thyroid hormones in two selected examples. MAJOR CONCLUSIONS In the intestinal epithelium and the retina, TRα1 and TRβ2 are expressed at the level of the precursors where they induce cell proliferation and differentiation, respectively. Moreover, these different functions result from the integration of the hormone signal with other intrinsic pathways, which play a fundamental role in progenitor/stem cell physiology. GENERAL SIGNIFICANCE Taken together, the interaction of TRs with other signaling pathways, specifically in stem/progenitor cells, is a new concept that may have biological relevance in therapeutic approaches aimed to target stem cells such as tissue engineering and cancer. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
Affiliation(s)
- Maria Sirakov
- Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, Belgium
| | | | | | | |
Collapse
|
34
|
GATA-4/-6 and HNF-1/-4 families of transcription factors control the transcriptional regulation of the murine Muc5ac mucin during stomach development and in epithelial cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:869-76. [DOI: 10.1016/j.bbagrm.2012.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/13/2012] [Accepted: 04/17/2012] [Indexed: 02/07/2023]
|
35
|
Arvanitis DN, Davy A. Regulation and misregulation of Eph/ephrin expression. Cell Adh Migr 2012; 6:131-7. [PMID: 22568953 DOI: 10.4161/cam.19690] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The erythropoietin-producing hepatocellular (Eph) receptors form the largest family of receptor tyrosine kinases. Upon interaction of the Eph receptors with their ligands the ephrins, signaling cascades are initiated downstream of both receptor and ligand, a feature known as bidirectional signaling. The Eph receptors and ephrin ligands mediate important roles in embryonic development, particularly in establishing tissue organization by mediating cell adhesion or cell repulsion. In several adult tissues, at least one Eph/ephrin pair is found to play critical roles in tissue physiology and homeostasis. In recent years numerous members of this family have gained considerable attention since changes in their expression levels are a typical feature in cancer cells. Despite the fact that Eph/ephrin developmental expression profiles are well documented, little is known on transcriptional and post-transcriptional mechanisms that permits their highly specific, graded, complementary or overlapping expression patterns. Therefore understanding the transcriptional and post-transcriptional mechanisms regulating Eph/ephrin expression has far-reaching significance in biology. This review provides an overview of the mechanisms regulating Eph/ephrin expression. We highlight important emerging mechanisms of Eph/ephrin regulation or misregulation such as epigenetics and miRNAs.
Collapse
Affiliation(s)
- Dina N Arvanitis
- Centre de Biologie du Développement, CNRS, Université de Toulouse, Toulouse, France
| | | |
Collapse
|
36
|
Stockinger S, Hornef MW, Chassin C. Establishment of intestinal homeostasis during the neonatal period. Cell Mol Life Sci 2011; 68:3699-712. [PMID: 21952827 PMCID: PMC11114965 DOI: 10.1007/s00018-011-0831-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 09/07/2011] [Accepted: 09/07/2011] [Indexed: 12/15/2022]
Abstract
The intestinal mucosa faces the challenge of regulating the balance between immune tolerance towards commensal bacteria, environmental stimuli and food antigens on the one hand, and induction of efficient immune responses against invading pathogens on the other hand. This regulatory task is of critical importance to prevent inappropriate immune activation that may otherwise lead to chronic inflammation, tissue disruption and organ dysfunction. The most striking example for the efficacy of the adaptive nature of the intestinal mucosa is birth. Whereas the body surfaces are protected from environmental and microbial exposure during fetal life, bacterial colonization and contact with potent immunostimulatory substances start immediately after birth. In the present review, we summarize the current knowledge on the mechanisms underlying the transition of the intestinal mucosa during the neonatal period leading to the establishment of a stable, life-long host-microbial homeostasis. The environmental exposure and microbial colonization during the neonatal period, and also the influence of maternal milk on the immune protection of the mucosa and the role of antimicrobial peptides, are described. We further highlight the molecular mechanisms of innate immune tolerance in neonatal intestinal epithelium. Finally, we link the described immunoregulatory mechanisms to the increased susceptibility to inflammatory and infectious diseases during the neonatal period.
Collapse
Affiliation(s)
- Silvia Stockinger
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| | - Mathias W. Hornef
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| | - Cécilia Chassin
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
37
|
Niu HJ, Chen X, Wang BM. Significance of expression of Sox2 and Cdx2 in gastric intestinal metaplasia. Shijie Huaren Xiaohua Zazhi 2011; 19:1295-1299. [DOI: 10.11569/wcjd.v19.i12.1295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the significance of expression of sex determining region Y-box 2 (Sox2) and caudal type homeobox transcription factor 2 (Cdx2) in gastric intestinal metaplasia (IM).
METHODS: The expression of SOX2 and CDX2 proteins in 80 paraffin-embedded specimens of gastritis and mild/moderate/severe IM was detected by immunohistochemistry. The mRNA levels of Sox2 and Cdx2 in 40 endoscopic biopsy specimens of gastritis and mild/moderate/severe IM were quantified by real-time Q-PCR.
RESULTS: Both SOX2 and CDX2 proteins were located in the nuclei of normal gastric and normal intestinal epithelial cells. The positive rates of SOX2 and CDX2 protein expression in gastritis and mild/moderate/severe IM were 94.4% and 5.6%, 75.0% and 50.0%, 23.5% and 85.7%, and 9.5% and 90.5%, respectively (all P < 0.05). The relative expression levels of Sox2 and Cdx2 mRNAs in gastritis and mild/moderate/severe IM were 0.5778 ± 0.0778 and 0.0517 ± 0.0218, 0.1496 ± 0.0384 and 0.1402 ± 0.0300, and 0.1131 ± 0.0384 and 0.3453 ± 0.0537, respectively (all P < 0.05). The levels of Sox2 transcripts decreased but those of Cdx2 transcripts increased with the progression of IM. There is an inverse correlation between the expression levels of Sox2 and Cdx2 (r < 0).
CONCLUSION: Down-regulation of Sox2 and ectopic expression of Cdx2 were found in the progression of gastric IM.
Collapse
|