1
|
Xu S, Yu Z, Li Z, Wang Z, Shi C, Li J, Wang F, Liu H. Wheat bran inclusion level impacts its net energy by shaping gut microbiota and regulating heat production in gestating sows. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:45-57. [PMID: 37779510 PMCID: PMC10539868 DOI: 10.1016/j.aninu.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/15/2023] [Accepted: 06/25/2023] [Indexed: 10/03/2023]
Abstract
An accurate estimation of net energy (NE) of wheat bran is essential for precision feeding of sows. However, the effects of inclusion level on NE of wheat bran have not been reported. Inclusion level was hypothesized to impact NE of wheat bran by regulating gut microbiota and partitioning of heat production. Therefore, twelve multiparous sows (Yorkshire × Landrace; 2 to 4 parity) were assigned to a replicated 3 × 6 Youden square with 3 successive periods and 6 diets in each square. The experiment included a corn-soybean meal diet (WB0) and five diets including 9.8% (WB10), 19.5% (WB20), 29.2% (WB30), 39.0% (WB40) and 48.7% wheat bran (WB50), respectively. Each period included 6 d of adaptation to diets followed by 6 d for heat production measurement using open-circuit respiration chambers. Compared with other groups, WB30, WB40, and WB50 enriched different fiber-degrading bacteria genera (P < 0.05). Apparent total tract digestibility of neutral detergent fiber and acid detergent fiber of wheat bran were greater in WB30 and WB40 (P < 0.05). Physical activity (standing and sitting) decreased as inclusion level increased (P = 0.04), which tended to decrease related heat production (P = 0.07). Thermic effect of feeding (TEF) was higher in WB50 than other treatments (P < 0.01). Metabolizable energy of wheat bran was similar among treatment groups (except for WB10). NE of wheat bran conformed to a quadratic regression equation with inclusion level (R2 = 0.99, P < 0.01) and peaked at an inclusion level of 35.3%. In conclusion, increasing inclusion level decreased energy expenditure of sows on physical activity and promoted growth of fiber-degrading bacteria, which improved energy utilization of fiber. Fermentation of wheat bran fiber by Prevotellaceae_UCG-003 and norank_f__Paludibacteraceae might increase TEF. Consequently, sows utilized energy in wheat bran most efficiently at an inclusion level of 35.3%.
Collapse
Affiliation(s)
- Song Xu
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Zirou Yu
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Zongliang Li
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Zijie Wang
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Chenyu Shi
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jian Li
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Fenglai Wang
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Hu Liu
- State Key Lab of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| |
Collapse
|
2
|
Chen Y, Liang Y, Cao L, Dong X, Sun D. Neuroendocrine differentiation: a risk fellow in colorectal cancer. World J Surg Oncol 2023; 21:89. [PMID: 36899368 PMCID: PMC9999536 DOI: 10.1186/s12957-023-02952-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/14/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Neuroendocrine differentiation (NED) is often found in colorectal cancer (CRC) and may have unique biological behavior, which has not been previously delineated. Here, we explore the relationship between CRC, NED, and clinicopathological factors. We also offer a preliminary explanation of the mechanism underlying the malignant biological behavior of NED in CRC. METHODS Between 2013 and 2015, 394 CRC patients who underwent radical operations were selected for analysis. The relationship between NED and clinicopathological factors was analyzed. To further clarify the pivotal role of NED in CRC, we performed bioinformatic analyses and identified genes that may be involved in NED, which were obtained from in silico data from The Cancer Genome Atlas (TCGA) database. Then, we conducted functional enrichment analyses and confirmed the critical pathways for intensive study. Moreover, we detected the expression of key proteins by immunohistochemistry and analyzed the correlation of their expression with NED. RESULTS The statistical analysis showed that CRC with NED was positively correlated with lymph node metastasis. Through bioinformatic analysis, we found that chromogranin A (CgA) was positively correlated with invasion and lymph node metastasis. ErbB2 and PIK3R1, which are key proteins in the PI3K-Akt signaling pathway, were closely related to NED. Furthermore, we determined that the PI3K-Akt signaling pathway likely plays a critical role in the NED of CRC. CONCLUSIONS CRC with NED is associated with lymph node metastasis. The PI3K-Akt signaling pathway, which is closely related to CRC, may be the mechanism promoting the malignant biological behavior of CRC with NED.
Collapse
Affiliation(s)
- Yue Chen
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Yu Liang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Lianqun Cao
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Xinxin Dong
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Deyu Sun
- Department of Radiation Oncology Gastrointestinal and Urinary and Musculoskeletal Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.
| |
Collapse
|
3
|
Li R, Rao JN, Smith AD, Chung HK, Xiao L, Wang JY, Turner DJ. miR-542-5p targets c-myc and negates the cell proliferation effect of SphK1 in intestinal epithelial cells. Am J Physiol Cell Physiol 2023; 324:C565-C572. [PMID: 36622069 PMCID: PMC9942902 DOI: 10.1152/ajpcell.00145.2022] [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: 04/04/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/10/2023]
Abstract
Intestinal epithelial barrier defects occur commonly during a variety of pathological conditions, though their underlying mechanisms are not completely understood. Sphingosine-1-phosphate (S1P) has been shown to be a critical regulator of proliferation and of maintenance of an intact intestinal epithelial barrier, as is also sphingosine kinase 1 (SphK1), the rate-limiting enzyme for S1P synthesis. SphK1 has been shown to modulate its effect on intestinal epithelial proliferation through increased levels of c-myc. We conducted genome-wide profile analysis to search for differential microRNA expression related to overexpressed SphK1 demonstrating adjusted expression of microRNA 542-5p (miR-542-5p). Here, we show that miR-542-5p is regulated by SphK1 activity and is an effector of c-myc translation that ultimately serves as a critical regulator of the intestinal epithelial barrier. miR-542-5p directly regulates c-myc translation through direct binding to the c-myc mRNA. Exogenous S1P analogs administered in vivo protect murine intestinal barrier from damage due to mesenteric ischemia reperfusion, and damaged intestinal tissue had increased levels of miR-542-5p. These results indicate that miR-542-5p plays a critical role in the regulation of S1P-mediated intestinal barrier function, and may highlight a novel role in potential therapies.
Collapse
Affiliation(s)
- Ruiyun Li
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore VA Medical Center, Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore VA Medical Center, Baltimore, Maryland
| | - Alexis D Smith
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore VA Medical Center, Baltimore, Maryland
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore VA Medical Center, Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore VA Medical Center, Baltimore, Maryland
- Cell Biology Group, Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Douglas J Turner
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore VA Medical Center, Baltimore, Maryland
| |
Collapse
|
4
|
Voß H, Moritz M, Pelczar P, Gagliani N, Huber S, Nippert V, Schlüter H, Hahn J. Tissue Sampling and Homogenization with NIRL Enables Spatially Resolved Cell Layer Specific Proteomic Analysis of the Murine Intestine. Int J Mol Sci 2022; 23:ijms23116132. [PMID: 35682811 PMCID: PMC9181169 DOI: 10.3390/ijms23116132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
For investigating the molecular physiology and pathophysiology in organs, the most exact data should be obtained; if not, organ-specific cell lines are analyzed, or the whole organ is homogenized, followed by the analysis of its biomolecules. However, if the morphological organization of the organ can be addressed, then, in the best case, the composition of molecules in single cells of the target organ can be analyzed. Laser capture microdissection (LCM) is a technique which enables the selection of specific cells of a tissue for further analysis of their molecules. However, LCM is a time-consuming two-dimensional technique, and optimal results are only obtained if the tissue is fixed, e.g., by formalin. Especially for proteome analysis, formalin fixation reduced the number of identifiable proteins, and this is an additional drawback. Recently, it was demonstrated that sampling of fresh-frozen (non-fixed) tissue with an infrared-laser is giving higher yields with respect to the absolute protein amount and number of identifiable proteins than conventional mechanical homogenization of tissues. In this study, the applicability of the infrared laser tissue sampling for the proteome analysis of different cell layers of murine intestine was investigated, using LC–MS/MS-based differential quantitative bottom-up proteomics. By laser ablation, eight consecutive layers of colon tissue were obtained and analyzed. However, a clear distinguishability of protein profiles between ascending, descending, and transversal colon was made, and we identified the different intestinal-cell-layer proteins, which are cell-specific, as confirmed by data from the Human Protein Atlas. Thus, for the first time, sampling directly from intact fresh-frozen tissue with three-dimensional resolution is giving access to the different proteomes of different cell layers of colon tissue.
Collapse
Affiliation(s)
- Hannah Voß
- Section/Core Facility Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (H.V.); (M.M.); (V.N.)
| | - Manuela Moritz
- Section/Core Facility Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (H.V.); (M.M.); (V.N.)
| | - Penelope Pelczar
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (P.P.); (N.G.); (S.H.)
| | - Nicola Gagliani
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (P.P.); (N.G.); (S.H.)
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (P.P.); (N.G.); (S.H.)
| | - Vivien Nippert
- Section/Core Facility Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (H.V.); (M.M.); (V.N.)
| | - Hartmut Schlüter
- Section/Core Facility Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (H.V.); (M.M.); (V.N.)
- Correspondence: (H.S.); (J.H.); Tel.: +49-1575-6085997 (H.S.); +49-1522-2827168 (J.H.)
| | - Jan Hahn
- Section/Core Facility Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20246 Hamburg, Germany; (H.V.); (M.M.); (V.N.)
- Correspondence: (H.S.); (J.H.); Tel.: +49-1575-6085997 (H.S.); +49-1522-2827168 (J.H.)
| |
Collapse
|
5
|
Dias R, Bergamo P, Maurano F, Rotondi Aufiero V, Luongo D, Mazzarella G, Bessa-Pereira C, Pérez-Gregorio M, Rossi M, Freitas V. First morphological-level insights into the efficiency of green tea catechins and grape seed procyanidins on a transgenic mouse model of celiac disease enteropathy. Food Funct 2021; 12:5903-5912. [PMID: 34028481 DOI: 10.1039/d1fo01263k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alternative or complementary treatments to a gluten-free diet are urgently needed for Celiac Disease. By exploiting the health-promoting properties of polyphenols on a transgenic mouse model of Celiac Disease enteropathy, this study provides the first in vivo evidence regarding the ability of 1 mg day-1 doses of green tea catechins and grape seed procyanidins to ameliorate some of the most characteristic histological changes of gliadin-treated DQ8 mice, including villus flattening, crypt hyperplasia, and infiltration of intraepithelial lymphocytes. Mechanistically, polyphenols were found to increase the intestinal nucleophilic tone of DQ8 mice by orchestrating an adaptive antioxidant response characterized by enhanced GSR enzyme activity and GSH content. Taken together, this work constitutes a highly relevant breakthrough as it provides the fundamental basis concerning the significance of natural polyphenols to be used in, for instance, the development of innovative functional foods aimed at CD individuals.
Collapse
Affiliation(s)
- Ricardo Dias
- QUINOA-LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto, Portugal.
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | - Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | - Catarina Bessa-Pereira
- QUINOA-LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto, Portugal.
| | - Maria Pérez-Gregorio
- QUINOA-LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto, Portugal.
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy and European Laboratory for the Investigation of Food-Induced Diseases (ELFID), Avellino, Italy
| | - Victor Freitas
- QUINOA-LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto, Portugal.
| |
Collapse
|
6
|
Review: Importance of colostrum supply and milk feeding intensity on gastrointestinal and systemic development in calves. Animal 2020; 14:s133-s143. [PMID: 32024575 DOI: 10.1017/s1751731119003148] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Feeding management of the postnatal and preweaning calf has an important impact on calf growth and development during this critical period and affects the health and well-being of the calves. After birth, an immediate and sufficient colostrum supply is a prerequisite for successful calf rearing. Colostrum provides high amounts of nutrient as well as non-nutrient factors that promote the immune system and intestinal maturation of the calf. The maturation and function of the neonatal intestine enable the calf to digest and absorb the nutrients provided by colostrum and milk. Therefore, colostrum intake supports the start of anabolic processes in several tissues, stimulating postnatal body growth and organ development. After the colostrum feeding period, an intensive milk feeding protocol, that is, at least 20% of BW milk intake/day, is required to realise the calf potential for growth and organ development during the preweaning period. Insufficient milk intake delays postnatal growth and may have detrimental effects on organ development, for example, the intestine and the mammary gland. The somatotropic axis as the main postnatal endocrine regulatory system for body growth is stimulated by the intake of high amounts of colostrum and milk and indicates the promotion of anabolic metabolism in calves. The development of the forestomach is an important issue during the preweaning period in calves, and forestomach maturation is best achieved by solid feed intake. Unfortunately, intensive milk-feeding programmes compromise solid feed intake during the first weeks of life. In the more natural situation for beef calves, when milk and solid feed intake occurs at the same time, calves benefit from the high milk intake as evidenced by enhanced body growth and organ maturation without impaired forestomach development during weaning. To realise an intensive milk-feeding programme, it is recommended that the weaning process should not start too early and that solid feed intake should be at a high extent despite intensive milk feeding. A feeding concept based on intensive milk feeding prevents hunger and abnormal behaviour of the calves and fits the principles of animal welfare during preweaning calf rearing. Studies on milk performance in dairy cows indicate that feeding management during early calf rearing influences lifetime performance. Therefore, an intensive milk-feeding programme affects immediate as well as long-term performance, probably by programming metabolic pathways during the preweaning period.
Collapse
|
7
|
Reese AT, Carmody RN. Thinking Outside the Cereal Box: Noncarbohydrate Routes for Dietary Manipulation of the Gut Microbiota. Appl Environ Microbiol 2019; 85:e02246-18. [PMID: 30504210 PMCID: PMC6498178 DOI: 10.1128/aem.02246-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota is a diverse and dynamic ecological community that is increasingly recognized to play important roles in host metabolic, immunological, and behavioral functioning. As such, identifying new routes for manipulating the microbiota may provide valuable additional methods for improving host health. Dietary manipulations and prebiotic supplementation are active targets of research for altering the microbiota, but to date, this work has disproportionately focused on carbohydrates. However, many other resources can limit or shape microbial growth. Here, we provide a brief overview of the resource landscape in the mammalian gut and review relevant literature documenting associations between noncarbohydrate nutrients and the composition of the gut microbiota. To spur future work and accelerate translational applications, we propose that researchers take new approaches for studying the effects of diet on gut microbial communities, including more-careful consideration of media for in vitro experiments, measurement of absolute as well as relative abundances, concerted efforts to articulate how physiology may differ between humans and the animal models used in translational studies, and leveraging natural variation for additional insights. Finally, we close with a discussion of how to determine when or where to employ these potential dietary levers for manipulating the microbiota.
Collapse
Affiliation(s)
- Aspen T Reese
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
- Society of Fellows, Harvard University, Cambridge, Massachusetts, USA
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
8
|
Sun X, Zhu MJ. Butyrate Inhibits Indices of Colorectal Carcinogenesis via Enhancing α-Ketoglutarate-Dependent DNA Demethylation of Mismatch Repair Genes. Mol Nutr Food Res 2018; 62:e1700932. [PMID: 29577594 DOI: 10.1002/mnfr.201700932] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/28/2018] [Indexed: 12/16/2022]
Abstract
SCOPE Butyrate, the fermentation end product of gut microbiota in the colon, is known for its antitumor effects, but the mechanisms remained to be defined. α-ketoglutarate (α-KG) mediates DNA demethylation and aberrant epigenetic modifications are associated with carcinogenesis. The objectives of this study are to evaluate the effects of butyrate on α-KG mediated epigenetic modification in colorectal adenocarcinoma HT-29 and Caco-2 cells. METHODS AND RESULTS Butyrate suppressed proliferation, potentiated differentiation, and induced apoptosis in both HT-29 and Caco-2 cells, associated with enhanced expression of isocitrate dehydrogenase 1 (IDH1) and pyruvate dehydrogenase. Furthermore, butyrate upregulated acetyl-CoA and α-KG, concomitant with enhanced histone acetylation and DNA demethylation in the promoter of DNA mismatch repair (MMR) gene. Knocking down IDH1 abolished the positive effects of butyrate on CRC apoptosis and MMR protein expression, in conjunction with reduced α-KG content. Importantly, α-KG supplementation recovered the beneficial effects of butyrate in IDH1-deficient cells. CONCLUSION In summary, butyrate inhibits indices of colorectal carcinogenesis in an α-KG-dependent manner.
Collapse
Affiliation(s)
- Xiaofei Sun
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| |
Collapse
|
9
|
Kerr BJ, Trabue SL, Andersen DS. Narasin effects on energy, nutrient, and fiber digestibility in corn-soybean meal or corn-soybean meal-dried distillers grains with solubles diets fed to 16-, 92-, and 141-kg pigs1. J Anim Sci 2017. [DOI: 10.2527/jas.2017.1732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
10
|
Cross sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of IBD patients. Sci Rep 2017; 7:9523. [PMID: 28842640 PMCID: PMC5573342 DOI: 10.1038/s41598-017-10034-5] [Citation(s) in RCA: 247] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract of uncertain origin, which includes ulcerative colitis (UC) and Crohn’s disease (CD). The composition of gut microbiota may change in IBD affected individuals, but whether dysbiosis is the cause or the consequence of inflammatory processes in the intestinal tissue is still unclear. Here, the composition of the microbiota and the metabolites in stool of 183 subjects (82 UC, 50 CD, and 51 healthy controls) were determined. The metabolites content and the microbiological profiles were significantly different between IBD and healthy subjects. In the IBD group, Firmicutes, Proteobacteria, Verrucomicrobia, and Fusobacteria were significantly increased, whereas Bacteroidetes and Cyanobacteria were decreased. At genus level Escherichia, Faecalibacterium, Streptococcus, Sutterella and Veillonella were increased, whereas Bacteroides, Flavobacterium, and Oscillospira decreased. Various metabolites including biogenic amines, amino acids, lipids, were significantly increased in IBD, while others, such as two B group vitamins, were decreased in IBD compared to healthy subjects. This study underlines the potential role of an inter-omics approach in understanding the metabolic pathways involved in IBD. The combined evaluation of metabolites and fecal microbiome can be useful to discriminate between healthy subjects and patients with IBD.
Collapse
|
11
|
Chen Y, Liu F, Meng Q, Ma S. Is neuroendocrine differentiation a prognostic factor in poorly differentiated colorectal cancer? World J Surg Oncol 2017; 15:71. [PMID: 28351413 PMCID: PMC5370452 DOI: 10.1186/s12957-017-1139-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/23/2017] [Indexed: 01/08/2023] Open
Abstract
Background To determine the prognostic relevance of neuroendocrine differentiation in poorly differentiated colorectal cancer. Methods The clinicopathological features and survival of 70 patients with poorly differentiated colorectal cancer were analyzed retrospectively. Chromogranin A and synaptophysin were used as neuroendocrine markers. Patients were followed-up for more than 3 years or until death. Results Of these 70 patients, 36 showed neuroendocrine differentiation. In univariate prognostic analysis, the patients with lymph node metastasis (P < 0.001), advanced TNM stage (P < 0.001), and neuroendocrine differentiation (P = 0.003) tended to have a poor prognosis. However, only lymph node metastasis was associated with a poor prognosis in multivariate analysis (P < 0.001). Patients with neuroendocrine differentiation were associated with lymph node metastasis (P = 0.006). Conclusions Neuroendocrine differentiation in poorly differentiated colorectal cancer was not a direct prognostic factor in these patients. Lymph node metastasis was a direct prognostic factor in these patients. Patients with neuroendocrine differentiation were associated with lymph node metastasis.
Collapse
Affiliation(s)
- Yue Chen
- Department of colorectal surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, People's Republic of China
| | - Fang Liu
- Department of colorectal surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, People's Republic of China.
| | - Qingkai Meng
- Department of colorectal surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, People's Republic of China
| | - Siping Ma
- Department of colorectal surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, People's Republic of China
| |
Collapse
|
12
|
Yang X, Jiang Z, Gong Y, Zheng C, Hu Y, Wang L, Huang L, Ma X. Supplementation of pre-weaning diet with l-arginine has carry-over effect to improve intestinal development in young piglets. CANADIAN JOURNAL OF ANIMAL SCIENCE 2016. [DOI: 10.1139/cjas-2015-0043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study tested the hypothesis that pre-weaning supplemental arginine may have a carry-over effect on intestinal growth and development of piglets immediately after weaning. Fifty-four [Duroc × (Landrace × Yorkshire)] piglets were fed a milk replacer diet supplemented with 0 (control), 4, or 8 g kg−1 of l-arginine from d 4 to 21 of age (6 replicate pens of 3 piglets per group). Piglets were then weaned to a common corn–soybean meal diet and fed for another 21 d. On day 42, 6 pigs per treatment were randomly selected for blood and tissue sampling. Arginine supplementation improved body weight of the piglets on d 42, average daily gain during d 22–31 (P < 0.05). Supplementation of 8 g kg−1 arginine decreased feed:gain (F:G) ratio in piglets during d 22–31 (P = 0.010). Compared with controls, 8 g kg−1 arginine improved villous height in duodenum, jejunum, and ileum; villous area in duodenum and jejunum; relative intestine weight; and plasma contents of insulin at d 42 (P < 0.05). Arginine supplementation increased mucosal protein content in all 3 segments of the small intestine (P < 0.05). These novel results clearly demonstrate a carry-over effect of pre-weaning supplementation with arginine on enhanced intestinal growth and development in the early post-weaning period.
Collapse
Affiliation(s)
- X.F. Yang
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - Z.Y. Jiang
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - Y.L. Gong
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - C.T. Zheng
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - Y.J. Hu
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - L. Wang
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - L. Huang
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| | - X.Y. Ma
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
- State Laboratory of Animal and Poultry Breeding, Ministry of Agriculture; Key Laboratory of Animal Nutrition and Feed Science (South China); Guangdong Public Laboratory of Animal Breeding and Nutrition; Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, People’s Republic of China
| |
Collapse
|
13
|
Chung HK, Chen Y, Rao JN, Liu L, Xiao L, Turner DJ, Yang P, Gorospe M, Wang JY. Transgenic Expression of miR-222 Disrupts Intestinal Epithelial Regeneration by Targeting Multiple Genes Including Frizzled-7. Mol Med 2015; 21:676-687. [PMID: 26252186 DOI: 10.2119/molmed.2015.00147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/30/2015] [Indexed: 01/15/2023] Open
Abstract
Defects in intestinal epithelial integrity occur commonly in various pathologies. miR-222 is implicated in many aspects of cellular function and plays an important role in several diseases, but its exact biological function in the intestinal epithelium is underexplored. We generated mice with intestinal epithelial tissue-specific overexpression of miR-222 to investigate the function of miR-222 in intestinal physiology and diseases in vivo. Transgenic expression of miR-222 inhibited mucosal growth and increased susceptibility to apoptosis in the small intestine, thus leading to mucosal atrophy. The miR-222-elevated intestinal epithelium was vulnerable to pathological stress, since local overexpression of miR-222 not only delayed mucosal repair after ischemia/reperfusion-induced injury, but also exacerbated gut barrier dysfunction induced by exposure to cecal ligation and puncture. miR-222 overexpression also decreased expression of the Wnt receptor Frizzled-7 (FZD7), cyclin-dependent kinase 4 and tight junctions in the mucosal tissue. Mechanistically, we identified the Fzd7 messenger ribonucleic acid (mRNA) as a novel target of miR-222 and found that [miR-222/Fzd7 mRNA] association repressed Fzd7 mRNA translation. These results implicate miR-222 as a negative regulator of normal intestinal epithelial regeneration and protection by downregulating expression of multiple genes including the Fzd7. Our findings also suggest a novel role of increased miR-222 in the pathogenesis of mucosal growth inhibition, delayed healing and barrier dysfunction.
Collapse
Affiliation(s)
- Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Yu Chen
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Lan Liu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Douglas J Turner
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging (NIA)-Intramural Research Program (IRP), National Institutes of Health, Baltimore, Maryland, United States of America
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
14
|
Huang Q, Su YB, Li DF, Liu L, Huang CF, Zhu ZP, Lai CH. Effects of inclusion levels of wheat bran and body weight on ileal and fecal digestibility in growing pigs. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2015; 28:847-54. [PMID: 25925062 PMCID: PMC4412981 DOI: 10.5713/ajas.14.0769] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/23/2014] [Accepted: 01/09/2015] [Indexed: 11/27/2022]
Abstract
The objective of this study was to determine the effects of graded inclusions of wheat bran (0%, 9.65%, 48.25% wheat bran) and two growth stages (from 32.5 to 47.2 kg and 59.4 to 78.7 kg, respectively) on the apparent ileal digestibility (AID), apparent total tract digestibility (ATTD) and hindgut fermentation of nutrients and energy in growing pigs. Six light pigs (initial body weight [BW] 32.5±2.1 kg) and six heavy pigs (initial BW 59.4±3.2 kg) were surgically prepared with a T-cannula in the distal ileum. A difference method was used to calculate the nutrient and energy digestibility of wheat bran by means of comparison with a basal diet consisting of corn-soybean meal (0% wheat bran). Two additional diets were formulated by replacing 9.65% and 48.25% wheat bran by the basal diet, respectively. Each group of pigs was allotted to a 6×3 Youden square design, and pigs were fed to three experimental diets during three 11-d periods. Hindgut fermentation values were calculated as the differences between ATTD and AID values. For the wheat bran diets, the AID and ATTD of dry matter (DM), ash, organic matter (OM), carbohydrates (CHO), gross energy (GE), and digestible energy (DE) decreased with increasing inclusion levels of wheat bran (p<0.05). While only AID of CHO and ATTD of DM, ash, OM, CHO, GE, and DE content differed (p<0.05) when considering the BW effect. For the wheat bran ingredient, there was a wider variation effect (p<0.01) on the nutrient and energy digestibility of wheat bran in 9.65% inclusion level due to the coefficient of variation (CV) of the nutrient and energy digestibility being higher at 9.65% compared to 48.25% inclusion level of wheat bran. Digestible energy content of wheat bran at 48.25% inclusion level (4.8 and 6.7 MJ/kg of DM, respectively) fermented by hindgut was significantly higher (p<0.05) than that in 9.65% wheat bran inclusion level (2.56 and 2.12 MJ/kg of DM, respectively), which was also affected (p<0.05) by two growth stages. This increase in hindgut fermentation caused the difference in ileal DE (p<0.05) to disappear at total tract level. All in all, increasing wheat bran levels in diets negatively influences the digestibility of some nutrients in pigs, while it positively affects the DE fermentation in the hindgut.
Collapse
Affiliation(s)
| | | | | | | | | | - Z. P. Zhu
- The New Hope Liu He Co., Ltd, 610063, Chengdu, China
| | - C. H. Lai
- Corresponding Author: C. H. Lai. Tel: +86-10-6273-2734, Fax: +86-10-6273-3688, E-mail:
| |
Collapse
|
15
|
Linking our understanding of mammary gland metabolism to amino acid nutrition. Amino Acids 2014; 46:2447-62. [DOI: 10.1007/s00726-014-1818-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/25/2014] [Indexed: 12/15/2022]
|
16
|
Bolick DT, Chen T, O. Alves LA, Tong Y, Wu D, Joyner LT, Oriá RB, Guerrant RL, Fu Z. Intestinal cell kinase is a novel participant in intestinal cell signaling responses to protein malnutrition. PLoS One 2014; 9:e106902. [PMID: 25184386 PMCID: PMC4153720 DOI: 10.1371/journal.pone.0106902] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 08/08/2014] [Indexed: 01/17/2023] Open
Abstract
Nutritional deficiency and stress can severely impair intestinal architecture, integrity and host immune defense, leading to increased susceptibility to infection and cancer. Although the intestine has an inherent capability to adapt to environmental stress, the molecular mechanisms by which the intestine senses and responds to malnutrition are not completely understood. We hereby report that intestinal cell kinase (ICK), a highly conserved serine/threonine protein kinase, is a novel component of the adaptive cell signaling responses to protein malnutrition in murine small intestine. Using an experimental mouse model, we demonstrated that intestinal ICK protein level was markedly and transiently elevated upon protein deprivation, concomitant with activation of prominent pro-proliferation and pro-survival pathways of Wnt/β-catenin, mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK), and protein kinase B (PKB/Akt) as well as increased expression of intestinal stem cell markers. Using the human ileocecal epithelial cell line HCT-8 as an invitro model, we further demonstrated that serum starvation was able to induce up-regulation of ICK protein in intestinal epithelial cells in a reversible manner, and that serum albumin partially contributed to this effect. Knockdown of ICK expression in HCT-8 cells significantly impaired cell proliferation and down-regulated active β-catenin signal. Furthermore, reduced ICK expression in HCT-8 cells induced apoptosis through a caspase-dependent mechanism. Taken together, our findings suggest that increased ICK expression/activity in response to protein deprivation likely provides a novel protective mechanism to limit apoptosis and support compensatory mucosal growth under nutritional stress.
Collapse
Affiliation(s)
- David T. Bolick
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tufeng Chen
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Luís Antonio O. Alves
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
| | - Yixin Tong
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
- Gastrointestinal Surgery Center, Tongji Hospital, Huazhong University of Science & Technology, Hubei, China
| | - Di Wu
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
| | - Linwood T. Joyner
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
| | - Reinaldo B. Oriá
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
| | - Richard L. Guerrant
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (ZF); (RLG)
| | - Zheng Fu
- Department of Medicine, Center for Global Health, Digestive Research Center of Excellence, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (ZF); (RLG)
| |
Collapse
|
17
|
Liu L, Christodoulou-Vafeiadou E, Rao JN, Zou T, Xiao L, Chung HK, Yang H, Gorospe M, Kontoyiannis D, Wang JY. RNA-binding protein HuR promotes growth of small intestinal mucosa by activating the Wnt signaling pathway. Mol Biol Cell 2014; 25:3308-18. [PMID: 25165135 PMCID: PMC4214778 DOI: 10.1091/mbc.e14-03-0853] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Inhibition of growth of the intestinal epithelium, a rapidly self-renewing tissue, is commonly found in various critical disorders. The RNA-binding protein HuR is highly expressed in the gut mucosa and modulates the stability and translation of target mRNAs, but its exact biological function in the intestinal epithelium remains unclear. Here, we investigated the role of HuR in intestinal homeostasis using a genetic model and further defined its target mRNAs. Targeted deletion of HuR in intestinal epithelial cells caused significant mucosal atrophy in the small intestine, as indicated by decreased cell proliferation within the crypts and subsequent shrinkages of crypts and villi. In addition, the HuR-deficient intestinal epithelium also displayed decreased regenerative potential of crypt progenitors after exposure to irradiation. HuR deficiency decreased expression of the Wnt coreceptor LDL receptor-related protein 6 (LRP6) in the mucosal tissues. At the molecular level, HuR was found to bind the Lrp6 mRNA via its 3'-untranslated region and enhanced LRP6 expression by stabilizing Lrp6 mRNA and stimulating its translation. These results indicate that HuR is essential for normal mucosal growth in the small intestine by altering Wnt signals through up-regulation of LRP6 expression and highlight a novel role of HuR deficiency in the pathogenesis of intestinal mucosal atrophy under pathological conditions.
Collapse
Affiliation(s)
- Lan Liu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201
| | | | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Tongtong Zou
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Hong Yang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | | | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Veterans Affairs Medical Center, Baltimore, MD 21201; Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
18
|
Huang C, Guo Y, Yuan J. Dietary taurine impairs intestinal growth and mucosal structure of broiler chickens by increasing toxic bile acid concentrations in the intestine. Poult Sci 2014; 93:1475-83. [PMID: 24879697 DOI: 10.3382/ps.2013-03533] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Three experiments were conducted to determine the effect of taurine on the intestinal development, bile acid concentrations, and hormonal status of chickens. In experiment 1, a total of 250 one-day-old broilers were randomly allocated to 5 treatments and supplemented with 0, 0.25, 0.50, 1.00, and 2.00 g/kg of taurine, respectively. Growth performance, weight and length of the small intestine, and intestinal morphology were measured on d 7, 22, and 44. The gene expression levels of several hormones, including epidermal growth factor and cholecystokinin, were also evaluated. In experiment 2, 60 one-day-old broilers were supplemented with 0, 1.0, and 5.0 g/kg of taurine to assess cell proliferation in the jenunal crypt. In experiment 3, 100 newly hatched broilers were assigned randomly to 5 treatments (0, 0.10, 0.50, 2.00, 8.00 g/kg of taurine) to evaluate the bile acid concentrations in the jejunal mucosa. Our results indicated that dietary taurine decreased the length and weight of small intestine, the villus width, surface area, and crypt depth in the duodenum and jejunum (P < 0.05). Taurine also increased the expression of cholecystokinin and epidermal growth factor on the jejunal mucosa (P < 0.001). Taurine has little effect on stimulating the proliferation of intestinal crypt cells, except for 5 g/kg of taurine supplementation on d 14 (P < 0.05). Additionally, a linear increase in the jejunal concentrations of taurocholic acid, taurochenodeoxycholic acid, and taurolithocholic acid was observed on d 7 in broilers fed increasing levels of taurine. In conclusion, we suggested that taurine impairs intestinal mucosal development partly through generation of toxic bile acids.
Collapse
Affiliation(s)
- Chunxi Huang
- State Key Laboratory for Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Yuming Guo
- State Key Laboratory for Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Jianmin Yuan
- State Key Laboratory for Animal Nutrition, China Agricultural University, Beijing 100193, China
| |
Collapse
|
19
|
Erlotinib promotes endoplasmic reticulum stress-mediated injury in the intestinal epithelium. Toxicol Appl Pharmacol 2014; 278:45-52. [PMID: 24768708 DOI: 10.1016/j.taap.2014.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/10/2014] [Accepted: 04/15/2014] [Indexed: 01/23/2023]
Abstract
Erlotinib, a popular drug for treating non-small cell lung cancer (NSCLC), causes diarrhea in approximately 55% of patients receiving this drug. In the present study, we found that erlotinib induced barrier dysfunction in rat small intestine epithelial cells (IEC-6) by increasing epithelial permeability and down-regulating E-cadherin. The mRNA levels of various pro-inflammatory cytokines (Il-6, Il-25 and Il-17f) were increased after erlotinib treatment in IEC-6 cells. Erlotinib concentration- and time-dependently induced apoptosis and endoplasmic reticulum (ER) stress in both IEC-6 and human colon epithelial cells (CCD 841 CoN). Intestinal epithelial injury was also observed in male C57BL/6J mice administrated with erlotinib. Knockdown of C/EBP homologous protein (CHOP) with small interference RNA partially reversed erlotinib-induced apoptosis, production of IL-6 and down-regulation of E-cadherin in cultured intestinal epithelial cells. In conclusion, erlotinib caused ER stress-mediated injury in the intestinal epithelium, contributing to its side effects of diarrhea in patients.
Collapse
|
20
|
Chung HK, Rao JN, Zou T, Liu L, Xiao L, Gu H, Turner DJ, Yang P, Wang JY. Jnk2 deletion disrupts intestinal mucosal homeostasis and maturation by differentially modulating RNA-binding proteins HuR and CUGBP1. Am J Physiol Cell Physiol 2014; 306:C1167-75. [PMID: 24740539 DOI: 10.1152/ajpcell.00093.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Homeostasis and maturation of the mammalian intestinal epithelium are preserved through strict regulation of cell proliferation, apoptosis, and differentiation, but the exact mechanism underlying this process remains largely unknown. c-Jun NH2-terminal kinase 2 (JNK2) is highly expressed in the intestinal mucosa, and its activation plays an important role in proliferation and also mediates apoptosis in cultured intestinal epithelial cells (IECs). Here, we investigated the in vivo function of JNK2 in the regulation of intestinal epithelial homeostasis and maturation by using a targeted gene deletion approach. Targeted deletion of the jnk2 gene increased cell proliferation within the crypts in the small intestine and disrupted mucosal maturation as indicated by decreases in the height of villi and the villus-to-crypt ratio. JNK2 deletion also decreased susceptibility of the intestinal epithelium to apoptosis. JNK2-deficient intestinal epithelium was associated with an increase in the level of the RNA-binding protein HuR and with a decrease in the abundance of CUG-binding protein 1 (CUGBP1). In studies in vitro, JNK2 silencing protected intestinal epithelial cell-6 (IEC-6) cells against apoptosis and this protection was prevented by inhibiting HuR. Ectopic overexpression of CUGBP1 repressed IEC-6 cell proliferation, whereas CUGBP1 silencing enhanced cell growth. These results indicate that JNK2 is essential for maintenance of normal intestinal epithelial homeostasis and maturation under biological conditions by differentially modulating HuR and CUGBP1.
Collapse
Affiliation(s)
- Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Tongtong Zou
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Lan Liu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Hui Gu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Douglas J Turner
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland; and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
21
|
Chen G, Qiu Y, Sun L, Yu M, Wang W, Xiao W, Yang Y, Liu Y, Yang S, Teitelbaum DH, Ma Y, Lu D, Yang H. The jagged-2/notch-1/hes-1 pathway is involved in intestinal epithelium regeneration after intestinal ischemia-reperfusion injury. PLoS One 2013; 8:e76274. [PMID: 24098462 PMCID: PMC3789708 DOI: 10.1371/journal.pone.0076274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/26/2013] [Indexed: 12/27/2022] Open
Abstract
Background Notch signaling plays a critical role in the maintenance of intestinal crypt epithelial cell proliferation. The aim of this study was to investigate the role of Notch signaling in the proliferation and regeneration of intestinal epithelium after intestinal ischemia reperfusion (I/R) injury. Methods Male Sprague-Dawley rats were subjected to sham operation or I/R by occlusion of the superior mesenteric artery (SMA) for 20 min. Intestinal tissue samples were collected at 0, 1, 2, 4, and 6 h after reperfusion. Proliferation of the intestinal epithelium was evaluated by immunohistochemical staining of proliferating nuclear antigen (PCNA). The mRNA and protein expression levels of Notch signaling components were examined using Real-time PCR and Western blot analyses. Immunofluorescence was also performed to detect the expression and location of Jagged-2, cleaved Notch-1, and Hes-1 in the intestine. Finally, the γ-secretase inhibitor DAPT and the siRNA for Jagged-2 and Hes-1 were applied to investigate the functional role of Notch signaling in the proliferation of intestinal epithelial cells in an in vitro IEC-6 culture system. Results I/R injury caused increased intestinal crypt epithelial cell proliferation and increased mRNA and protein expression of Jagged-2, Notch-1, and Hes-1. The immunofluorescence results further confirmed increased protein expression of Jagged-2, cleaved Notch-1, and Hes-1 in the intestinal crypts. The inhibition of Notch signaling with DAPT and the suppression of Jagged-2 and Hes-1 expression using siRNA both significantly inhibited the proliferation of IEC-6 cells. Conclusion The Jagged-2/Notch-1/Hes-1 signaling pathway is involved in intestinal epithelium regeneration early after I/R injury by increasing crypt epithelial cell proliferation.
Collapse
Affiliation(s)
- Guoqing Chen
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yang Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yong Liu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Songwei Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Daniel H. Teitelbaum
- Department of Surgery, the University of Michigan Medical School, Ann Arbor, Michigan
| | - Yuanhang Ma
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Dingsong Lu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
22
|
Xiao L, Rao JN, Zou T, Liu L, Cao S, Martindale JL, Su W, Chung HK, Gorospe M, Wang JY. miR-29b represses intestinal mucosal growth by inhibiting translation of cyclin-dependent kinase 2. Mol Biol Cell 2013; 24:3038-46. [PMID: 23904268 PMCID: PMC3784378 DOI: 10.1091/mbc.e13-05-0287] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The epithelium of the intestinal mucosa is a rapidly self-renewing tissue in the body, and defects in the renewal process occur commonly in various disorders. miR-29b functions as a biological repressor of normal intestinal mucosal growth by repressing CDK2 translation through direct interaction with its mRNA, representing a novel therapeutic target for patients with mucosal atrophy. The epithelium of the intestinal mucosa is a rapidly self-renewing tissue in the body, and defects in the renewal process occur commonly in various disorders. microRNAs (miRNAs) posttranscriptionally regulate gene expression and are implicated in many aspects of cellular physiology. Here we investigate the role of miRNA-29b (miR-29b) in the regulation of normal intestinal mucosal growth and further validate its target mRNAs. miRNA expression profiling studies reveal that growth inhibition of the small intestinal mucosa is associated with increased expression of numerous miRNAs, including miR-29b. The simple systemic delivery of locked nucleic acid–modified, anti–miR-29b-reduced endogenous miR-29b levels in the small intestinal mucosa increases cyclin-dependent kinase 2 (CDK2) expression and stimulates mucosal growth. In contrast, overexpression of the miR-29b precursor in intestinal epithelial cells represses CDK2 expression and results in growth arrest in G1 phase. miR-29b represses CDK2 translation through direct interaction with the cdk2 mRNA via its 3′-untranslated region (3′-UTR), whereas point mutation of miR-29b binding site in the cdk2 3′-UTR prevents miR-29b–induced repression of CDK2 translation. These results indicate that miR-29b inhibits intestinal mucosal growth by repressing CDK2 translation.
Collapse
Affiliation(s)
- Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201 Laboratory of Genetics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Feng L, Peng Y, Wu P, Hu K, Jiang WD, Liu Y, Jiang J, Li SH, Zhou XQ. Threonine affects intestinal function, protein synthesis and gene expression of TOR in Jian carp (Cyprinus carpio var. Jian). PLoS One 2013; 8:e69974. [PMID: 23922879 PMCID: PMC3724917 DOI: 10.1371/journal.pone.0069974] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 06/14/2013] [Indexed: 01/24/2023] Open
Abstract
This study aimed to investigate the effects of threonine (Thr) on the digestive and absorptive ability, proliferation and differentiation of enterocytes, and gene expression of juvenile Jian carp (Cyprinus carpio var. Jian). First, seven isonitrogenous diets containing graded levels of Thr (7.4-25.2 g/kg diet) were fed to the fishes for 60 days. Second, enterocyte proliferation and differentiation were assayed by culturing enterocytes with graded levels of Thr (0-275 mg/l) in vitro. Finally, enterocytes were cultured with 0 and 205 mg/l Thr to determine protein synthesis. The percent weight gain (PWG), specific growth rate, feed intake, feed efficiency, protein retention value, activities of trypsin, lipase and amylase, weights and protein contents of hepatopancreas and intestine, folds heights, activities of alkaline phosphatase (AKP), γ- glutamyl transpeptidase and Na(+)/K(+)-ATPase in all intestinal segments, glutamate-oxaloacetate transaminase (GOT) and glutamate-pyruvate transaminase (GPT) activities in hepatopancreas, and 4E-BP2 gene expression in muscle, hepatopancreas and intestinal segments were significantly enhanced by Thr (p<0.05). However, the plasma ammonia concentration and TOR gene expression decreased (p<0.05). In vitro, Thr supplement significantly increased cell numbers, protein content, the activities of GOT, GPT, AKP and Na(+)/K(+)-ATPase, and protein synthesis rate of enterocytes, and decreased LDH activity and ammonia content in cell medium (p<0.05). In conclusion, Thr improved growth, digestive and absorptive capacity, enterocyte proliferation and differentiation, and protein synthesis and regulated TOR and 4E-BP2 gene expression in juvenile Jian carp. The dietary Thr requirement of juvenile Jian carp was 16.25 g/kg diet (51.3 g/kg protein) based on quadratic regression analysis of PWG.
Collapse
Affiliation(s)
- Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Peng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kai Hu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shu-Hong Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
24
|
Perrone EE, Liu L, Turner DJ, Strauch ED. Bile salts increase epithelial cell proliferation through HuR-induced c-Myc expression. J Surg Res 2012; 178:155-64. [PMID: 22626558 DOI: 10.1016/j.jss.2012.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 01/05/2012] [Accepted: 02/16/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Bile salts increase intestinal mucosal proliferation through an increase in c-Myc, a transcription factor that controls the expression of numerous translation regulatory proteins. HuR is an RNA-binding protein that regulates translation of target mRNAs. RNA-binding proteins can control mRNA stability by binding to AU- and U-rich elements located in the 3'-untranslated regions (3'-UTRs) of target mRNAs. AIM To determine how bile salt-induced c-Myc stimulates enterocyte proliferation. METHODS Enterocyte proliferation was measured both in vivo using C57Bl6 mice and in vitro using IEC-6 cells after taurodeoxycholate (TDCA) supplementation. HuR and c-Myc protein expression was determined by immunoblot. c-Myc mRNA expression was determined by PCR. HuR expression was inhibited using specific small interfering RNA. HuR binding to c-Myc mRNA was determined by immunoprecipitation. RESULTS TDCA increased enterocyte proliferation in vivo and in vitro. TDCA stimulates translocation of HuR from the nucleus to the cytoplasm. Cytoplasmic HuR regulates c-Myc translation by HuR binding to the 3'-UTR of c-Myc mRNA. Increased TDCA-induced c-Myc increases enterocyte proliferation. CONCLUSIONS Bile salts have beneficial effects on the intestinal epithelial mucosa, which are important in maintaining intestinal mucosal integrity and function. These data further support an important beneficial role of bile salts in regulation of mucosal growth and repair. Decreased enterocyte exposure to luminal bile salts, as occurs during critical illness, liver failure, starvation, and intestinal injury, may have a detrimental effect on mucosal integrity.
Collapse
Affiliation(s)
- Erin E Perrone
- Department of Pediatric Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
25
|
|
26
|
Bodiga VL, Bodiga S, Surampudi S, Boindala S, Putcha U, Nagalla B, Subramaniam K, Manchala R. Effect of vitamin supplementation on cisplatin-induced intestinal epithelial cell apoptosis in Wistar/NIN rats. Nutrition 2011; 28:572-80. [PMID: 22189195 DOI: 10.1016/j.nut.2011.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 08/17/2011] [Accepted: 09/07/2011] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Chemotherapeutic agents induce small intestinal mucositis that is characterized structurally by crypt loss and villus atrophy and functionally by absorptive and barrier impairments. We studied the effect of selected individual vitamins and multiple-vitamin mixture supplementation in modulating cisplatin-induced intestinal damage and apoptosis. METHODS Thirty-six male Wistar/NIN rats 20 wk old and fed the control diet (AIN-93G) were randomly divided into six groups. Five groups were administered cisplatin (2.61 mg/kg of body weight) once a week for 3 wk and were concomitantly provided the control diet or riboflavin, folate, α- tocopherol, or a multiple-vitamin mixture supplemented diet. The sixth group served as a control for cisplatin and received saline as the vehicle. Intestinal epithelial cell apoptosis was monitored by morphometry, M30 staining, DNA fragmentation, and caspase-3 activity. Functional and structural integrities were determined by measuring activities of alkaline phosphatase and lysine ala-dipeptidyl aminopeptidase and the villus height/crypt depth ratio. Oxidative burden was assessed as the formation of thiobarbituric acid-reactive substances and protein carbonyls. Plasma levels of selected vitamins were also measured. RESULTS Cisplatin administration significantly increased intestinal apoptosis in the villus and crypt regions that correlated with increased oxidative damage, decreased Bcl-2/Bax, and compromised functional integrity. Riboflavin, folate, and the multiple-vitamin mixture supplementation attenuated the cisplatin-induced increase in apoptotic indices, with a decrease in oxidative burden, increased Bcl-2/Bax, and improved functional and structural integrities. The α-tocopherol supplementation, although effective in attenuating oxidative stress and improving functional integrity, failed to lower the apoptotic indices. CONCLUSIONS Riboflavin, folate, and the multiple-vitamin supplementation proved to be more efficacious in attenuating the cisplatin-induced intestinal damage and associated changes in apoptosis.
Collapse
Affiliation(s)
- Vijaya Lakshmi Bodiga
- Pathology Division, National Institute of Nutrition, Hyderabad, Andhra Pradesh, India.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Zahiri HR, Perrone EE, Strauch ED. Bile salt supplementation acts via the farnesoid X receptor to alleviate lipopolysaccharide-induced intestinal injury. Surgery 2011; 150:480-9. [DOI: 10.1016/j.surg.2011.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Accepted: 07/06/2011] [Indexed: 01/16/2023]
|
28
|
Iwashita Y, Sakiyama T, Musch MW, Ropeleski MJ, Tsubouchi H, Chang EB. Polyamines mediate glutamine-dependent induction of the intestinal epithelial heat shock response. Am J Physiol Gastrointest Liver Physiol 2011; 301:G181-7. [PMID: 21512157 PMCID: PMC3129932 DOI: 10.1152/ajpgi.00054.2011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Heat shock proteins (Hsps) are highly conserved proteins that play a role in cytoprotection and maintaining intestinal homeostasis. Glutamine is essential for the optimal induction of intestinal epithelial Hsp expression, but its mechanisms of action are incompletely understood. Glutamine is a substrate for polyamine synthesis and stimulates the activity of ornithine decarboxylase (ODC), a key enzyme for polyamine synthesis, in intestinal epithelial cells. Thus we investigated whether polyamines (putrescine, spermidine, or spermine) and their precursor ornithine mediate the induction of Hsp expression in IEC-18 rat intestinal epithelial cells. As previously observed, glutamine was required for heat stress induction of Hsp70 and Hsp25, although it had little effect under basal conditions. Under conditions of glutamine depletion, supplementation of ornithine or polyamines restored the heat-induced expression of Hsp70 and Hsp25. When ODC was inhibited by α-difluoromethylornithine (DFMO), an irreversible ODC inhibitor, the heat stress induction of Hsp70 and Hsp25 was decreased significantly, even in the presence of glutamine. Ornithine, polyamines, and DFMO did not modify the nuclear localization of heat shock transcription factor 1 (HSF-1). However, DFMO dramatically reduced glutamine-dependent HSF-1 binding to an oligonucleotide with heat shock elements (HSE), which was increased by glutamine. In addition, exogenous polyamines recovered the DNA-binding activity. These results indicate that polyamines play a critical role in the glutamine-dependent induction of the intestinal epithelial heat shock response through facilitation of HSF-1 binding to HSE.
Collapse
Affiliation(s)
- Yuji Iwashita
- 1Department of Digestive and Life-style related Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Toshio Sakiyama
- 1Department of Digestive and Life-style related Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mark W. Musch
- 2Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Mark J. Ropeleski
- 3Gastrointestinal Diseases Research Unit, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Hirohito Tsubouchi
- 1Department of Digestive and Life-style related Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Eugene B. Chang
- 2Department of Medicine, University of Chicago, Chicago, Illinois; and
| |
Collapse
|
29
|
Affiliation(s)
- Rao N. Jaladanki
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| | - Jian-Ying Wang
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| |
Collapse
|
30
|
De Conto C, Oevermann A, Burgener IA, Doherr MG, Blum JW. Gastrointestinal tract mucosal histomorphometry and epithelial cell proliferation and apoptosis in neonatal and adult dogs. J Anim Sci 2010; 88:2255-64. [DOI: 10.2527/jas.2009-2511] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
31
|
Abstract
The gastrointestinal tract (GIT) is a metabolically significant site of sulfur amino acid (SAA) metabolism in the body and metabolises about 20 % of the dietary methionine intake which is mainly transmethylated to homocysteine and trans-sulfurated to cysteine. The GIT accounts for about 25 % of the whole-body transmethylation and trans-sulfuration. In addition, in vivo studies in young pigs indicate that the GIT is a site of net homocysteine release and thus may contribute to the homocysteinaemia. The gut also utilises 25 % of the dietary cysteine intake and the cysteine uptake by the gut represents about 65 % of the splanchnic first-pass uptake. Moreover, we recently showed that SAA deficiency significantly suppresses intestinal mucosal growth and reduces intestinal epithelial cell proliferation, and increases intestinal oxidant stress in piglets. These recent findings indicate that intestinal metabolism of dietary methionine and cysteine is nutritionally important for intestinal mucosal growth. Besides their role in protein synthesis, methionine and cysteine are precursors of important molecules. S-adenosylmethionine, a metabolite of methionine, is the principal biological methyl donor in mammalian cells and a precursor for polyamine synthesis. Cysteine is the rate-limiting amino acid for glutathione synthesis, the major cellular antioxidant in mammals. Further studies are warranted to establish how SAA metabolism regulates gut growth and intestinal function, and contributes to the development of gastrointestinal diseases. The present review discusses the evidence of SAA metabolism in the GIT and its functional and nutritional importance in gut function and diseases.
Collapse
|
32
|
García-Miranda P, Peral MJ, Ilundain AA. Rat small intestine expresses the reelin-Disabled-1 signalling pathway. Exp Physiol 2010; 95:498-507. [DOI: 10.1113/expphysiol.2009.050682] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
33
|
Xiao L, Rao JN, Zou T, Liu L, Yu TX, Zhu XY, Donahue JM, Wang JY. Induced ATF-2 represses CDK4 transcription through dimerization with JunD inhibiting intestinal epithelial cell growth after polyamine depletion. Am J Physiol Cell Physiol 2010; 298:C1226-34. [PMID: 20181929 DOI: 10.1152/ajpcell.00021.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intestinal epithelium is a rapidly self-renewing tissue in the body, and its homeostasis is tightly regulated by numerous factors including polyamines. Decreased levels of cellular polyamines increase activating transcription factor (ATF)-2, but the exact role and mechanism of induced ATF-2 in the regulation of intestinal epithelial cell (IEC) growth remain elusive. Cyclin-dependent kinase (CDK) 4 is necessary for the G1-to-S phase transition during the cell cycle, and its expression is predominantly controlled at the transcription level. Here, we reported that induced ATF-2 following polyamine depletion repressed CDK4 gene transcription in IECs by increasing formation of the ATF-2/JunD heterodimers. ATF-2 formed complexes with JunD as measured by immunoprecipitation using the ATF-2 and JunD antibodies and by glutathione S-transferase (GST) pull-down assays using GST-ATF-2 fusion proteins. Studies using various mutants of GST-ATF-2 revealed that formation of the ATF-2/JunD dimers depended on the COOH-terminal basic region-leucine zipper domain of ATF-2. Polyamine depletion increased ATF-2/JunD complex and inhibited CDK4 transcription as indicated by a decrease in the levels of CDK4-promoter activity and its mRNA. ATF-2 silencing not only prevented inhibition of CDK4 transcription in polyamine-deficient cells but also abolished repression of CDK4 expression induced by ectopic JunD overexpression. ATF-2 silencing also promoted IEC growth in polyamine-depleted cells. These results indicate that induced ATF-2/JunD association following polyamine depletion represses CDK4 transcription, thus contributing to the inhibition of IEC growth.
Collapse
Affiliation(s)
- Lan Xiao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
WU XUDONG, SUN LIXIN, ZHA WEIBIN, STUDER ELAINE, GURLEY EMILY, CHEN LI, WANG XUAN, HYLEMON PHILLIPB, PANDAK WILLIAMM, SANYAL ARUNJ, ZHANG LUYONG, WANG GUANGJI, CHEN JIE, WANG JIAN, ZHOU HUIPING. HIV protease inhibitors induce endoplasmic reticulum stress and disrupt barrier integrity in intestinal epithelial cells. Gastroenterology 2010; 138:197-209. [PMID: 19732776 PMCID: PMC4644065 DOI: 10.1053/j.gastro.2009.08.054] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 07/28/2009] [Accepted: 08/20/2009] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Human immunodeficiency virus (HIV) protease inhibitor (PI)-induced adverse effects have become a serious clinical problem. In addition to their metabolic and cardiovascular complications, these drugs also frequently cause severe gastrointestinal disorders, including mucosal erosions, epithelial barrier dysfunction, and diarrhea. However, the exact mechanisms underlying gastrointestinal adverse effects of HIV PIs remain unknown. This study investigated whether HIV PIs disrupt intestinal epithelial barrier integrity by activating endoplasmic reticulum (ER) stress. METHODS The most commonly used HIV PIs (lopinavir, ritonavir, and amprenavir) were used; their effects on ER stress activation and epithelial paracellular permeability were examined in vitro as well as in vivo using wild-type and CHOP(-)/(-) mice. RESULTS Treatment with lopinavir and ritonavir, but not amprenavir, induced ER stress, as indicated by a decrease in secreted alkaline phosphatase activities and an increase in the unfolded protein response. This activated ER stress partially impaired the epithelial barrier integrity by promoting intestinal epithelial cell apoptosis. CHOP silencing by specific small hairpin RNA prevented lopinavir- and ritonavir-induced barrier dysfunction in cultured intestinal epithelial cells, whereas CHOP(-)/(-) mice exhibited decreased mucosal injury after exposure to lopinavir and ritonavir. CONCLUSIONS HIV PIs induce ER stress and activate the unfolded protein response in intestinal epithelial cells, thus resulting in disruption of the epithelial barrier integrity.
Collapse
Affiliation(s)
- XUDONG WU
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - LIXIN SUN
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - WEIBIN ZHA
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - ELAINE STUDER
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - EMILY GURLEY
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - LI CHEN
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - XUAN WANG
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - PHILLIP B. HYLEMON
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia,Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia
| | - WILLIAM M. PANDAK
- Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia
| | - ARUN J. SANYAL
- Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia
| | - LUYONG ZHANG
- China Pharmaceutical University, Nanjing, Jiangsu, China
| | - GUANGJI WANG
- China Pharmaceutical University, Nanjing, Jiangsu, China
| | - JIE CHEN
- Department of Surgery, University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - JIAN–YING WANG
- Department of Surgery, University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - HUIPING ZHOU
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia,Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia,China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
35
|
Sphingosine-1-phosphate protects intestinal epithelial cells from apoptosis through the Akt signaling pathway. Dig Dis Sci 2009; 54:499-510. [PMID: 18654850 PMCID: PMC2696985 DOI: 10.1007/s10620-008-0393-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 06/17/2008] [Indexed: 12/09/2022]
Abstract
OBJECTIVE The regulation of apoptosis of intestinal mucosal cells is important in maintenance of normal intestinal physiology. SUMMARY Sphingosine-1-phosphate (S1P) has been shown to play a critical role in cellular protection to otherwise lethal stimuli in several nonintestinal tissues. METHODS The current study determines whether S1P protected normal intestinal epithelial cells (IECs) from apoptosis and whether Akt activation was the central pathway for this effect. RESULTS S1P demonstrated significantly reduced levels of apoptosis induced by tumor necrosis factor-alpha (TNF-alpha)/cycloheximide (CHX). S1P induced increased levels of phosphorylated Akt and increased Akt activity, but did not affect total amounts of Akt. This activation of Akt was associated with decreased levels of both caspase-3 protein levels and of caspase-3 activity. Inactivation of Akt by treatment with the PI3K chemical inhibitor LY294002 or by overexpression of the dominant negative mutant of Akt (DNMAkt) prevented the protective effect of S1P on apoptosis. Additionally, silencing of the S1P-1 receptor by specific siRNA demonstrated a lesser decrease in apoptosis to S1P exposure. CONCLUSION These results indicate that S1P protects intestinal epithelial cells from apoptosis via an Akt-dependent pathway.
Collapse
|
36
|
Robertson DJ, Sandler RS, Ahnen DJ, Greenberg ER, Mott LA, Cole BF, Baron JA. Gastrin, Helicobacter pylori, and colorectal adenomas. Clin Gastroenterol Hepatol 2009; 7:163-7. [PMID: 18929688 DOI: 10.1016/j.cgh.2008.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 08/12/2008] [Accepted: 09/07/2008] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Hypergastrinemia and Helicobacter pylori (Hp) infection have been associated with an increased risk for colorectal neoplasia in some studies. However, data from large prospective studies of both associations are lacking. The aim of this study was to evaluate whether serum gastrin levels and/or infection with Hp are associated with the subsequent development of colorectal adenomas. METHODS Subjects (all with a history of adenoma formation) were drawn from 2 previously completed adenoma chemoprevention trials. Participants underwent clearing colonoscopy at baseline with follow-up colonoscopy 1 and 4 years after enrollment. We used commercially available assays on fasting blood specimens to measure serum gastrin levels and Hp serologies 1 year after randomization. Risk ratios for adenoma and advanced adenoma development during the subsequent 3 years were computed by generalized linear regression. RESULTS Of the 1794 subjects randomized in the 2 trials, 685 had available serum and were included in the analyses. Gastrin levels were significantly higher in the 239 subjects with Hp titers indicating infection (mean, 88.3 pg/mL) than in those not infected (mean, 73.9 pg/mL; P < .001). In fully adjusted models, gastrin levels were not associated with incident adenoma development (risk ratio [RR], 1.10; 95% confidence interval [CI], 0.78-1.54) or advanced adenoma formation (RR, 0.82; 95% CI, 0.33-2.03). A positive Hp serology was associated with a decreased risk for adenoma formation (RR, 0.76; 95% CI, 0.60-0.96). CONCLUSIONS Neither hypergastrinemia nor serologic evidence of Hp infection were associated with an increased risk for recurrent adenoma development. These results do not support the notion that gastrin promotes colorectal carcinogenesis, at least at the stage of adenoma development.
Collapse
|
37
|
Flynn NE, Bird JG, Guthrie AS. Glucocorticoid regulation of amino acid and polyamine metabolism in the small intestine. Amino Acids 2008; 37:123-9. [DOI: 10.1007/s00726-008-0206-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2008] [Accepted: 11/01/2008] [Indexed: 10/21/2022]
|
38
|
Pisal DS, Yellepeddi VK, Kumar A, Palakurthi S. Transport of Surface Engineered Polyamidoamine (PAMAM) Dendrimers Across IPEC-J2 Cell Monolayers. Drug Deliv 2008; 15:515-22. [DOI: 10.1080/10717540802321826] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
39
|
Abstract
Gastric cancer is the second leading cause of cancer deaths worldwide. Although the link between Helicobacter pylori infection and gastric cancer is well established, little is known about the early development and detection of this malignant disease. Cancer is the disease of epithelia and recently, it has been suggested that some cancers originate in adult stem cells. Advances have been made in identifying the gastric epithelial stem cells and their immediate descendents, which act as progenitors giving rise to mucus-, acid-, pepsinogen-, and hormone-secreting cell lineages. Analyses of some genetically manipulated animal models in which the proliferation and differentiation program of the gastric stem/progenitor cells was altered by different approaches have provided some clues to the cellular origin of gastric cancer. Despite the challenges and the similarity between gastric epithelial progenitors and their differentiation program in mice and humans, it remains to be determined whether observations made in genetically engineered mice are also applicable to humans.
Collapse
Affiliation(s)
- Sherif M Karam
- Department of Anatomy, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates.
| |
Collapse
|
40
|
Kouch-el Filali M, Kilic E, Melis M, de Klein A, de Jong M, Luyten GPM. Expression of the SST receptor 2 in uveal melanoma is not a prognostic marker. Graefes Arch Clin Exp Ophthalmol 2008; 246:1585-92. [PMID: 18682975 DOI: 10.1007/s00417-008-0880-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Revised: 05/19/2008] [Accepted: 06/02/2008] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Uveal melanoma (UM) cells and neurohormone-producing cells both originate from the neural crest. Somatostatin receptors subtype 2 (SSTR2) are over-expressed in several tumors, often from neuroendocrine origin, and synthetic antagonists like octreotide and octreotate are being used as diagnostic or therapeutic agents. We investigated the SSTR2 expression in UM, and determined whether this expression was related to prognosis of the disease. MATERIALS AND METHODS UM cell lines and fresh primary UM samples were tested for SSTR2 expression by autoradiography (AR) using 125I-Tyr3-octreotate. Furthermore, UM cell lines were analyzed for SSTR2 mRNA expression with quantitative real-time RT-PCR. RESULTS Using AR, cell-surface SSTR2 expression was demonstrated in two UM metastatic cell lines, but no expression was detected in three cell lines derived from primary UM. However, all primary and metastatic UM cell lines showed mRNA expression levels for SSTR2 using quantitative real-time RT-PCR. Only three of 14 primary UM demonstrated moderate SSTR2 expression, and this expression was not significantly associated with tumor-free survival or any tested prognostic factor. CONCLUSIONS Based on the rare and low expression of SSTR2 found in primary UM specimens and in UM cell lines, we conclude that SSTR2 is not widely expressed in UM. Furthermore, SSTR2 expression was not associated with tumor-free survival and prognostic factors. Therefore SSTR2 is not suited as prognostic marker or therapeutic target in UM.
Collapse
|
41
|
Indigestible Material Attenuated Changes in Apoptosis in the Fasted Rat Jejunal Mucosa. Exp Biol Med (Maywood) 2008; 233:310-6. [DOI: 10.3181/0708-rm-228] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that fasting induced apoptosis and decreased cell proliferation in the rat intestinal mucosa. The aim was to investigate the effect of expanded polystyrene as indigestible material on apoptosis and cell proliferation in rat small intestinal mucosa during fasting. Male SD rats were divided into 3 groups. The first group was fed with chow and water ad libitum. The second group fasted for 72 hrs. The third group was fasted for 24 hrs and was fed expanded polystyrene. Intestinal apoptosis was evaluated by percent fragmented DNA assay, terminal deoxynucleotidyl transferase–mediated dUDP-biotin nick end-labeling (TUNEL) staining, and caspase-3 assay. Cell proliferation was analyzed by 5-bromo-2′-deoxyuridine (5-BrdU) uptake. Truncal vagotomy was performed to evaluate a role of the central nervous system. In the 72-hr fasted rat, mucosal height of the rat jejunum was decreased to 73% of that in rats fed ad libitum, and this decrease was partly restored to 90% in rats fed expanded polystyrene. The fragmented DNA was increased in fasted rats (28.0%) when compared with that in rats fed ad libitum (2.6%). The increase in fragmented DNA in fasted rats was recovered by feeding them expanded polystyrene (8.3%). TUNEL staining confirmed this result. The effect of polystyrene on apoptosis was decreased by truncal vagotomy. Expression of cleaved caspase-3 was increased in fasted rats, which was then decreased by feeding of expanded polystyrene. In contrast to apoptosis, feeding of expanded polystyrene had no reconstructive effect on 5-BrdU uptake in the intestinal epithelium, which was decreased by fasting to 60% of that in rats fed ad libitum. In conclusion, feeding of indigestible material partly restored the decrease in intestinal mucosal length in the fasted rats through the apoptotic pathway without any influence on BrdU uptake. Further exploration focused on the mechanism of this effect of indigestible material is required.
Collapse
|
42
|
Otsubo T, Akiyama Y, Yanagihara K, Yuasa Y. SOX2 is frequently downregulated in gastric cancers and inhibits cell growth through cell-cycle arrest and apoptosis. Br J Cancer 2008; 98:824-31. [PMID: 18268498 PMCID: PMC2259184 DOI: 10.1038/sj.bjc.6604193] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
SOX transcription factors are essential for embryonic development and play critical roles in cell fate determination, differentiation and proliferation. We previously reported that the SOX2 protein is expressed in normal gastric mucosae but downregulated in some human gastric carcinomas. To clarify the roles of SOX2 in gastric carcinogenesis, we carried out functional characterisation of SOX2 in gastric epithelial cell lines. Exogenous expression of SOX2 suppressed cell proliferation in gastric epithelial cell lines. Flow cytometry analysis revealed that SOX2-overexpressing cells exhibited cell-cycle arrest and apoptosis. We found that SOX2-mediated cell-cycle arrest was associated with decreased levels of cyclin D1 and phosphorylated Rb, and an increased p27Kip1 level. These cells exhibited further characteristics of apoptosis, such as DNA laddering and caspase-3 activation. SOX2 hypermethylation signals were observed in some cultured and primary gastric cancers with no or weak SOX2 expression. Among the 52 patients with advanced gastric cancers, those with cancers showing SOX2 methylation had a significantly shorter survival time than those without this methylation (P=0.0062). Hence, SOX2 plays important roles in growth inhibition through cell-cycle arrest and apoptosis in gastric epithelial cells, and the loss of SOX2 expression may be related to gastric carcinogenesis and poor prognosis.
Collapse
Affiliation(s)
- T Otsubo
- Department of Molecular Oncology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45, Yushima, Tokyo 113-8519, Japan
| | | | | | | |
Collapse
|
43
|
Xiao L, Rao JN, Zou T, Liu L, Marasa BS, Chen J, Turner DJ, Zhou H, Gorospe M, Wang JY. Polyamines regulate the stability of activating transcription factor-2 mRNA through RNA-binding protein HuR in intestinal epithelial cells. Mol Biol Cell 2007; 18:4579-90. [PMID: 17804813 PMCID: PMC2043536 DOI: 10.1091/mbc.e07-07-0675] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Maintenance of intestinal mucosal epithelial integrity requires polyamines that modulate the expression of various genes involved in cell proliferation and apoptosis. Recently, polyamines were shown to regulate the subcellular localization of the RNA-binding protein HuR, which stabilizes its target transcripts such as nucleophosmin and p53 mRNAs. The activating transcription factor-2 (ATF-2) mRNA encodes a member of the ATF/CRE-binding protein family of transcription factors and was computationally predicted to be a target of HuR. Here, we show that polyamines negatively regulate ATF-2 expression posttranscriptionally and that polyamine depletion stabilizes ATF-2 mRNA by enhancing the interaction of the 3'-untranslated region (UTR) of ATF-2 with cytoplasmic HuR. Decreasing cellular polyamines by inhibiting ornithine decarboxylase (ODC) with alpha-difluoromethylornithine increased the levels of ATF-2 mRNA and protein, whereas increasing polyamines by ectopic ODC overexpression repressed ATF-2 expression. Polyamine depletion did not alter transcription via the ATF-2 gene promoter but increased the stability of ATF-2 mRNA. Increased cytoplasmic HuR in polyamine-deficient cells formed ribonucleoprotein complexes with the endogenous ATF-2 mRNA and specifically bound to 3'-UTR of ATF-2 mRNA on multiple nonoverlapping 3'-UTR segments. Adenovirus-mediated HuR overexpression elevated ATF-2 mRNA and protein levels, whereas HuR silencing rendered the ATF-2 mRNA unstable and prevented increases in ATF-2 mRNA and protein. Furthermore, inhibition of ATF-2 expression prevented the increased resistance of polyamine-deficient cells to apoptosis induced by treatment with tumor necrosis factor-alpha and cycloheximide. These results indicate that polyamines modulate the stability of ATF-2 mRNA by altering cytoplasmic HuR levels and that polyamine-modulated ATF-2 expression plays a critical role in regulating epithelial apoptosis.
Collapse
Affiliation(s)
- Lan Xiao
- *Cell Biology Group, Department of Surgery, and
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Jaladanki N. Rao
- *Cell Biology Group, Department of Surgery, and
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Tongtong Zou
- *Cell Biology Group, Department of Surgery, and
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Lan Liu
- *Cell Biology Group, Department of Surgery, and
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Bernard S. Marasa
- *Cell Biology Group, Department of Surgery, and
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Jie Chen
- *Cell Biology Group, Department of Surgery, and
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Douglas J. Turner
- *Cell Biology Group, Department of Surgery, and
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298; and
| | - Myriam Gorospe
- Laboratory of Cellular and Molecular Biology, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Jian-Ying Wang
- *Cell Biology Group, Department of Surgery, and
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| |
Collapse
|
44
|
Yellepeddi VK, Pisal DS, Kumar A, Kaushik RS, Hildreth MB, Guan X, Palakurthi S. Permeability of surface-modified polyamidoamine (PAMAM) dendrimers across Caco-2 cell monolayers. Int J Pharm 2007; 350:113-21. [PMID: 17913410 PMCID: PMC2266586 DOI: 10.1016/j.ijpharm.2007.08.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 08/17/2007] [Accepted: 08/21/2007] [Indexed: 11/25/2022]
Abstract
Aim of this study was to prepare polyamine-conjugated PAMAM dendrimers and study their permeability across Caco-2 cell monolayers. Polyamines, namely, arginine and ornithine were conjugated to the amine terminals of the G4 PAMAM dendrimers by Fmoc synthesis. The apical-to-basolateral (AB) and basolateral-to-apical (BA) apparent permeability coefficients (P(app)) for the PAMAM dendrimers increased by conjugating the dendrimers with both of the polyamines. The enhancement in permeability was dependent on the dendrimer concentration and duration of incubation. The correlation between monolayer permeability and the decrease in transepithelial electrical resistance (TEER) with both the PAMAM dendrimers and the polyamine-conjugated dendrimers suggests that paracellular transport is one of the mechanisms of transport across the epithelial cells. Cytotoxicity of the polyamine-conjugated dendrimers was evaluated in Caco-2 cells by MTT (methylthiazoletetrazolium) assay. Arginine-conjugated dendrimers were slightly more toxic than PAMAM dendrimer as well as ornithine-conjugated dendrimers. Though investigations on the possible involvement of other transport mechanisms are in progress, results of the present study suggest the potential of dendrimer-polyamine conjugates as drug carriers to increase the oral absorption of drugs.
Collapse
Affiliation(s)
- Venkata K. Yellepeddi
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Dipak S. Pisal
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Ajay Kumar
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Radhey S. Kaushik
- Department of Biology & Microbiology/Veterinary Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Michael B. Hildreth
- Department of Biology & Microbiology, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Xiangming Guan
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| |
Collapse
|
45
|
Zhang AH, Rao JN, Zou T, Liu L, Marasa BS, Xiao L, Chen J, Turner DJ, Wang JY. p53-Dependent NDRG1 expression induces inhibition of intestinal epithelial cell proliferation but not apoptosis after polyamine depletion. Am J Physiol Cell Physiol 2007; 293:C379-89. [PMID: 17442733 DOI: 10.1152/ajpcell.00547.2006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Normal intestinal mucosal growth requires polyamines that regulate expression of various genes involved in cell proliferation, growth arrest, and apoptosis. Our previous studies have shown that polyamine depletion stabilizes p53, resulting in inhibition of intestinal epithelial cell (IEC) proliferation, but the exact downstream targets of induced p53 are still unclear. The NDRG1 (N- myc downregulated gene-1) gene encodes a growth-related protein, and its transcription can be induced in response to stress. The current study tests the hypothesis that induced p53 inhibits IEC proliferation by upregulating NDRG1 expression following polyamine depletion. Depletion of cellular polyamines by inhibiting ornithine decarboxylase (ODC) with α-difluoromethylornithine not only induced p53 but also increased NDRG1 transcription as indicated by induction of the NDRG1 promoter activity and increased levels of NDRG1 mRNA and protein, all of which were prevented by using specific p53 siRNA and in cells with a targeted deletion of p53. In contrast, increased levels of cellular polyamines by ectopic expression of the ODC gene decreased p53 and repressed expression of NDRG1. Consistently, polyamine depletion-induced activation of the NDRG1-promoter was decreased when p53-binding sites within the NDRG1 proximal promoter region were deleted. Ectopic expression of the wild-type NDRG1 gene inhibited DNA synthesis and decreased final cell numbers regardless of the presence or absence of endogenous p53, whereas silencing NDRG1 promoted cell growth. However, overexpression of NDRG1 failed to directly induce cell death and to alter susceptibility to apoptosis induced by tumor necrosis factor-α/cycloheximide. These results indicate that NDRG1 is one of the direct mediators of induced p53 following polyamine depletion and that p53-dependent NDRG1 expression plays a critical role in the negative control of IEC proliferation.
Collapse
Affiliation(s)
- Ai-Hong Zhang
- Dept. of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The gastric epithelium is a complex structure formed into tubular branched gastric glands. The glands contain a wide variety of cell types concerned with the secretion of hydrochloric acid, proteases, mucus and a range of signalling molecules. All cell types originate from stem cells in the neck region of the gland, before migrating and differentiating to assume their characteristic positions and functions. Endocrine and local paracrine mediators are of crucial importance for maintaining structural and functional integrity of the epithelium, in the face of a hostile luminal environment. The first such mediator to be recognized, the hormone gastrin, was identified over a century ago and is now established as the major physiological stimulant of gastric acid secretion. Recent studies, including those using mice that overexpress or lack the gastrin gene, suggest a number of previously unrecognized roles for this hormone in the regulation of cellular proliferation, migration and differentiation. This review focuses on the identification of hitherto unsuspected gastrin-regulated genes and discusses the paracrine cascades that contribute to the maintenance of gastric epithelial architecture and secretory function. Helicobacter infection is also considered in cases where it shares targets and signalling mechanisms with gastrin.
Collapse
Affiliation(s)
- Rod Dimaline
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool L69 3BX, UK.
| | | |
Collapse
|
47
|
Turner DJ, Alaish SM, Zou T, Rao JN, Wang JY, Strauch ED. Bile salts induce resistance to apoptosis through NF-kappaB-mediated XIAP expression. Ann Surg 2007; 245:415-25. [PMID: 17435549 PMCID: PMC1877019 DOI: 10.1097/01.sla.0000236631.72698.99] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Apoptosis plays a critical role in intestinal mucosal homeostasis. We previously showed that the bile salt taurodeoxycholate has a beneficial effect on the intestinal mucosa through an increase in resistance to apoptosis mediated by nuclear factor (NF)-kappaB. The current study further characterizes the effect of bile salts on intestinal epithelial cell susceptibility to apoptosis and determines if the X-linked inhibitor of apoptosis protein (XIAP) regulates bile salt-induced resistance to apoptosis. Exposure of normal intestinal epithelial cells (IEC-6) to the conjugated bile salts taurodeoxycholate (TDCA) and taurochenodeoxycholate (TCDCA) resulted in an increase in resistance to tumor necrosis factor (TNF)-alpha and cycloheximide (CHX)-induced apoptosis, and NF-kappaB activation. Treatment with TDCA and TCDCA resulted in an increase in XIAP expression. Specific inhibition of NF-kappaB by infection with an adenoviral vector that expresses the IkappaBalpha super-repressor (IkappaBSR) prevented the induction of XIAP expression and the bile salt-mediated resistance to apoptosis. Treatment with the specific XIAP inhibitor Smac also overcame this increase in enterocyte resistance to apoptosis. Bile salts inhibited formation of the active caspase-3 from its precursor procaspase-3. Smac prevented the inhibitory effect of bile salts on caspase-3 activation. These results indicate that bile salts increase intestinal epithelial cell resistance to apoptosis through NF-kappaB-mediated XIAP expression. Bile salt-induced XIAP mediates resistance to TNF-alpha/CHX-induced apoptosis, at least partially, through inhibition of caspase-3 activity. These data support an important beneficial role of bile salts in regulation of mucosal integrity. Decreased enterocyte exposure to luminal bile salts, as occurs during starvation and parenteral nutrition, may have a detrimental effect on mucosal integrity.
Collapse
Affiliation(s)
- Douglas J Turner
- Departments of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Xiao L, Rao J, Zou T, Liu L, Marasa B, Chen J, Turner D, Passaniti A, Wang JY. Induced JunD in intestinal epithelial cells represses CDK4 transcription through its proximal promoter region following polyamine depletion. Biochem J 2007; 403:573-81. [PMID: 17253961 PMCID: PMC1876376 DOI: 10.1042/bj20061436] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 01/17/2007] [Accepted: 01/25/2007] [Indexed: 02/06/2023]
Abstract
Maintenance of intestinal epithelial integrity requires cellular polyamines that regulate expression of various genes involved in cell proliferation, growth arrest and apoptosis. In prior studies, depletion of cellular polyamines has been shown to stabilize JunD, a member of the AP-1 (activator protein-1) family of transcription factors, leading to inhibition of intestinal epithelial cell proliferation, but the exact downstream targets of induced JunD remain elusive. CDK4 (cyclin-dependent kinase 4) is essential for the G1- to S-phase transition during the cell cycle and its expression is primarily controlled at the transcriptional level. In the present study, we show that induced JunD in IECs (intestinal epithelial cells) is a transcriptional repressor of the CDK4 gene following polyamine depletion. Increased JunD in polyamine-deficient cells was associated with a significant inhibition of CDK4 transcription, as indicated by repression of CDK4-promoter activity and decreased levels of CDK4 mRNA and protein, all of which were prevented by using specific antisense JunD oligomers. Ectopic expression of the wild-type junD also repressed CDK4-promoter activity and decreased levels of CDK4 mRNA and protein without any effect on CDK2 expression. Gel shift and chromatin immunoprecipitation assays revealed that JunD bound to the proximal region of the CDK4-promoter in vitro as well as in vivo, while experiments using different CDK4-promoter mutants showed that transcriptional repression of CDK4 by JunD was mediated through an AP-1 binding site within this proximal sequence of the CDK4-promoter. These results indicate that induced JunD in IECs represses CDK4 transcription through its proximal promoter region following polyamine depletion.
Collapse
Key Words
- activator protein-1 (ap-1)
- α-difluoromethylornithine
- growth arrest
- intestinal epithelium
- ornithine decarboxylase
- transcriptional regulation
- ap-1, activator protein-1
- cdk, cyclin-dependent kinase
- chip, chromatin immunoprecipitation
- dfmo, α-difluoromethylornithine
- emsa, electrophoretic mobility-shift assay
- fbs, fetal bovine serum
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- iec, intestinal epithelial cell
- luc, luciferase
- pbs-t, pbs containing tween 20
- q-pcr, quantitative pcr
- rb, retinoblastoma tumour suppressor protein
- rt, reverse transcriptase
Collapse
Affiliation(s)
- Lan Xiao
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jaladanki N. Rao
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Tongtong Zou
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Lan Liu
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Bernard S. Marasa
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- †Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jie Chen
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Douglas J. Turner
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Antonino Passaniti
- †Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jian-Ying Wang
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- †Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| |
Collapse
|
49
|
Wang JY. Polyamines and mRNA stability in regulation of intestinal mucosal growth. Amino Acids 2007; 33:241-52. [PMID: 17404803 DOI: 10.1007/s00726-007-0518-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Accepted: 02/01/2007] [Indexed: 10/23/2022]
Abstract
The mammalian intestinal epithelium is a rapidly self-renewing tissue in the body, and its homeostasis is preserved through strict regulation of epithelial cell proliferation, growth arrest, and apoptosis. Polyamines are necessary for normal intestinal mucosal growth and decreasing cellular polyamines inhibits cell proliferation and disrupts epithelial integrity. An increasing body of evidence indicates that polyamines regulate intestinal epithelial cell renewal by virtue of their ability to modulate expression of various genes and that growth inhibition following polyamine depletion results primarily from the activation of growth-inhibiting genes rather than a simple decrease in expression of growth-promoting genes. In this review article, we will focus on changes in expression of growth-inhibiting genes following polyamine depletion and further analyze in some detail the mechanisms through which mRNA stability is regulated by RNA-binding proteins.
Collapse
Affiliation(s)
- J-Y Wang
- Cell Biology Group, Departments of Surgery and Pathology, University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA.
| |
Collapse
|
50
|
Abstract
Neurotensin (NT) is a brain-gut tridecapeptide that functions as a neurotransmitter/neuromodulator in the central nervous system (CNS) and as an endocrine agent in the periphery. NT has numerous physiologic effects on multiple organs. This review will focus on the effects of NT as a trophic factor for normal and neoplastic tissues. In this regard, NT may act as an endocrine agent or, in some instances, in a paracrine and/or autocrine fashion. These effects appear to be mediated predominantly through the G protein-coupled high-affinity NT receptor. However, some of the trophic effects may also be through the other two receptor subtypes, particularly the NT receptor type 3, which belongs to a recently identified family of sorting receptors. The signaling pathways mediating the effects of NT are multiple but most appear to activate the ERK signaling pathway, which then activates downstream transcription factors, ultimately leading to proliferation. NT may be a useful agent to enhance the growth of normal tissues such as the small bowel mucosa during periods of gut disuse or disease and, finally, the selective targeting of NT receptor subtypes on certain cancers may offer a novel strategy in the armamentarium of cancer chemotherapeutics.
Collapse
Affiliation(s)
- B Mark Evers
- Department of Surgery, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| |
Collapse
|