1
|
Almeida JZ, Vieira LA, Maside C, Ferreira ACA, Sá NAR, Correia HHV, Araújo VR, Raposo RS, Smitz J, Campello CC, Figueiredo JR, Oriá RB. In vitro cytotoxic effects of 5-Fluorouracil on isolated murine ovarian preantral follicles. Theriogenology 2022; 178:60-66. [PMID: 34775200 DOI: 10.1016/j.theriogenology.2021.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 12/01/2022]
Abstract
5 fluorouracil (5FU), an antineoplastic drug, is often utilized in the therapeutic regimen for several types of cancer, including the hepatoblastoma in children. The effects of 5FU on the population of ovarian preantral follicles, which is the largest oocyte reservoir, is still poorly understood. The integrity of the ovarian preantral follicle pool is important for lifelong fertility. The better understanding of such effects may favor intervention strategies to protect fertility in 5FU-treated children and women coping with cancer. To analyze the effects of 5FU on isolated murine secondary follicles in vitro, ovaries were collected from young mice (28-30 days old), and secondary follicles were isolated and cultured for 12 days in basic culture medium, with or without 5FU at concentrations of 0.3 mM, 1 mM, 3 mM, 10 mM, and 30 mM. In the in vitro study, we analyzed the percentage of morphologically normal follicles, antrum formation, follicular diameter, and hormone production. On day 12, oocytes were recovered for in vitro maturation. 5FU treatment did not alter the percentage of morphologically normal follicles. On day 12, only 1, 10, and 30 mM 5FU significantly reduced the percentage of antrum. From day 4 onwards, 5FU treatments significantly reduced follicle diameter. The meiosis resumption rate was significantly lower in all 5FU treatments. 5FU concentrations ≥3 mM reduced estradiol levels. In conclusion, 5FU does not affect follicular morphology. However, 5FU deleteriously affects follicular growth, estradiol production, and oocyte maturation in isolated ovarian follicles.
Collapse
Affiliation(s)
- Juliana Z Almeida
- Laboratory of Tissue Healing, Ontogeny, and Nutrition (LABICONTE), Department of Morphology, Institute of Biomedicine, School of Medicine, Federal University of Ceara (UFC), 1315 Rua Cel. Nunes de Melo, Fortaleza, CE, 60430-270, Brazil
| | - L A Vieira
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - C Maside
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - A C A Ferreira
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - N A R Sá
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - H H V Correia
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - V R Araújo
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - R S Raposo
- Experimental Biology Center, University of Fortaleza (UNIFOR), 1321 Av. Washington Soares, Fortaleza, CE, 60811-905, Brazil
| | - J Smitz
- Laboratory of Follicle Biology, Center for Reproductive Medicine, UZ Brussel, Laarbeeklaan 101, Brussels, B-1090, Brazil
| | - C C Campello
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil
| | - J R Figueiredo
- Faculty of Veterinary Medicine, Laboratory of Oocytes and Preantral Follicles Manipulation (LAMOFOPA), State University of Ceara (UECE), 1700 Av. Dr. Silas Munguba, Fortaleza, CE, 60741-000, Brazil.
| | - R B Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition (LABICONTE), Department of Morphology, Institute of Biomedicine, School of Medicine, Federal University of Ceara (UFC), 1315 Rua Cel. Nunes de Melo, Fortaleza, CE, 60430-270, Brazil
| |
Collapse
|
2
|
Molinari GS, Wojno M, McCracken VJ, Kwasek K. The use of dipeptide supplementation as a means of mitigating the negative effects of dietary soybean meal on Zebrafish Danio rerio. Comp Biochem Physiol A Mol Integr Physiol 2021; 257:110958. [PMID: 33865992 DOI: 10.1016/j.cbpa.2021.110958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022]
Abstract
Soybean meal (SBM) inclusion in aquaculture diets has been found to negatively affect growth and induce intestinal inflammation in fish. The objective of this study was to determine the effect of health-promoting dipeptide supplementation into SBM-based feeds on growth performance, intestinal health, and muscle free amino acid composition, an indicator of dietary amino acid availability, in a zebrafish model. There were five treatment groups in this study. The first group ((+) Control) received a fishmeal-based diet. The second group ((-) Control) received SBM-based diet. The last three groups (Ala-Glu, Car, and Ans) were fed SBM-based diets, supplemented with alanyl-glutamine, carnosine, and anserine respectively. The Ala-Glu and Car groups experienced a significantly higher weight gain than the (-) Control group, weighing 35.38% and 33.96% more, respectively at the conclusion of the study. There were no significant differences in gene expression among the groups, but Ala-Glu had the highest expression of both nutrient absorption genes measured, PepT1 and fabp2. Ala-Glu had significantly longer intestinal villi, and a significantly higher villus length-to-width ratio than the (-) Control group. The Car group had a significantly higher post-prandial tissue concentration of lysine, compared to the (-) Control group. The increase in villus surface area and expression of nutrient absorption genes represent an improvement in intestinal absorptive capacity in the Ala-Glu group. The results from this study provide support for the use of alanyl-glutamine and carnosine supplementation as a means of improving growth performance of zebrafish fed with a high level SBM-based diet.
Collapse
Affiliation(s)
- Giovanni S Molinari
- Center for Fisheries, Aquaculture, and Aquatic Sciences, Southern Illinois University, 1125 Lincoln Dr, Carbondale, IL 62901, USA
| | - Michal Wojno
- Center for Fisheries, Aquaculture, and Aquatic Sciences, Southern Illinois University, 1125 Lincoln Dr, Carbondale, IL 62901, USA
| | - Vance J McCracken
- Department of Biological Sciences, Southern Illinois University Edwardsville, 44 Circle Dr, Edwardsville, IL 62025, USA
| | - Karolina Kwasek
- Center for Fisheries, Aquaculture, and Aquatic Sciences, Southern Illinois University, 1125 Lincoln Dr, Carbondale, IL 62901, USA.
| |
Collapse
|
3
|
Almeida JZ, Lima LF, Vieira LA, Maside C, Ferreira ACA, Araújo VR, Duarte ABG, Raposo RS, Báo SN, Campello CC, Oliveira LFS, da Costa TP, Abreu JG, Figueiredo JR, Oriá RB. 5-Fluorouracil disrupts ovarian preantral follicles in young C57BL6J mice. Cancer Chemother Pharmacol 2021; 87:567-578. [PMID: 33471160 DOI: 10.1007/s00280-020-04217-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/11/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE 5-Fluorouracil (5-FU), an anti-cancer drug, has been used for hepatoblastoma (HB) chemotherapy in children, who may have impaired ovarian follicle pool reserve with lasting effects to reproduction. Therefore, this study aimed to investigate 5-FU effects on survival, growth, and morphology of ovarian preantral follicles from C57BL6J young mice. METHODS Experiments were carried-out both in vivo and in vitro. Mice were treated with 5-FU injection (450 mg/kg i.p) or saline and sacrificed 3 days after to obtain ovaries for histology and molecular biology. Ovaries for in vitro studies were obtained from unchallenged mice and cultured under basic culture medium (BCM) or BCM plus 5-FU (9.2, 46.1, 92.2 mM). Preantral follicles were classified according to developmental stages, and as normal or degenerated. To assess cell viability, caspase-3 immunostaining was performed. Transcriptional levels for apoptosis (Bax, Bcl2, p53, Bax/Bcl2) and Wnt pathway genes (Wnt2 and Wnt4) were also analyzed. Ultrastructural analyses were carried-out on non-cultured ovaries. In addition, β-catenin immunofluorescence was assessed in mouse ovaries. RESULTS The percentage of all-types normal follicles was significantly lower after 5-FU challenge. A total loss of secondary normal follicles was found in the 5-FU group. The highest 5-FU concentrations reduced the percentage of cultured normal primordial follicles. Large vacuoles were seen in granulosa cells and ooplasm of preantral follicles by electron microscopy. A significantly higher gene expression for Bax and Bax/Bcl2 ratio was seen after 5-FU treatment. A marked reduction in β-catenin immunolabeling was seen in 5-FU-challenged preantral follicles. In the in vitro experiments, apoptotic and Wnt gene transcriptions were significantly altered. CONCLUSION Altogether, our findings suggest that 5-FU can deleteriously affect the ovarian follicle reserve by reducing preantral follicles survival.
Collapse
Affiliation(s)
- Juliana Z Almeida
- Department of Morphology, Institute of Biomedicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, School of Medicine, Federal University of Ceara, 1315 Rua Cel. Nunes de Melo, Fortaleza, CE, 60430-270, Brazil
| | - Laritza F Lima
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Luís A Vieira
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Carolina Maside
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Anna C A Ferreira
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Valdevane R Araújo
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Ana B G Duarte
- Department of Morphology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Ramon S Raposo
- Experimental Biology Core, University of Fortaleza, Fortaleza, CE, Brazil
| | - Sônia N Báo
- Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil
| | - Cláudio C Campello
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Luiz F S Oliveira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thayse P da Costa
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Garcia Abreu
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José R Figueiredo
- Faculty of Veterinary Medicine, Laboratory of Manipulation of Oocytes and Preantral Follicles (LAMOFOPA), State University of Ceara, Fortaleza, CE, Brazil
| | - Reinaldo B Oriá
- Department of Morphology, Institute of Biomedicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, School of Medicine, Federal University of Ceara, 1315 Rua Cel. Nunes de Melo, Fortaleza, CE, 60430-270, Brazil.
| |
Collapse
|
4
|
Adikwu E, Biradee I, Ogungbaike T. Therapeutic benefit of resveratrol in 5-fluorouracil-induced nephrotoxicity in rats. BIOMEDICAL RESEARCH JOURNAL 2019. [DOI: 10.4103/bmrj.bmrj_19_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
5
|
Mattila J, Kokki K, Hietakangas V, Boutros M. Stem Cell Intrinsic Hexosamine Metabolism Regulates Intestinal Adaptation to Nutrient Content. Dev Cell 2018; 47:112-121.e3. [PMID: 30220570 PMCID: PMC6179903 DOI: 10.1016/j.devcel.2018.08.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 07/07/2018] [Accepted: 08/14/2018] [Indexed: 02/08/2023]
Abstract
The intestine is an organ with an exceptionally high rate of cell turnover, and perturbations in this process can lead to severe diseases such as cancer or intestinal atrophy. Nutrition has a profound impact on intestinal volume and cellular architecture. However, how intestinal homeostasis is maintained in fluctuating dietary conditions remains insufficiently understood. By utilizing the Drosophila midgut model, we reveal a novel stem cell intrinsic mechanism coupling cellular metabolism with stem cell extrinsic growth signal. Our results show that intestinal stem cells (ISCs) employ the hexosamine biosynthesis pathway (HBP) to monitor nutritional status. Elevated activity of HBP promotes Warburg effect-like metabolic reprogramming required for adjusting the ISC division rate according to nutrient content. Furthermore, HBP activity is an essential facilitator for insulin signaling-induced ISC proliferation. In conclusion, ISC intrinsic hexosamine synthesis regulates metabolic pathway activities and defines the stem cell responsiveness to niche-derived growth signals. HBP is a mediator of Drosophila midgut adaptation to nutrient content ISC intrinsic HBP is a necessary and sufficient driver of stem cell divisions HBP activity regulates a Warburg-like metabolic reprogramming of the intestine HBP activity determines the output of InR signaling of the ISCs
Collapse
Affiliation(s)
- Jaakko Mattila
- German Cancer Research Center, Division of Signaling and Functional Genomics and Heidelberg University, Heidelberg 69120, Germany
| | - Krista Kokki
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland; Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Ville Hietakangas
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland; Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Michael Boutros
- German Cancer Research Center, Division of Signaling and Functional Genomics and Heidelberg University, Heidelberg 69120, Germany.
| |
Collapse
|
6
|
Cavalcante PA, Prata MMG, Medeiros PHQS, Alves da Silva AV, Quetz JS, Reyes MAV, Rodrigues TS, Santos AKS, Ribeiro SA, Veras HN, Bona MD, Amaral MSMG, Rodrigues FAP, Lima IFN, Havt A, Lima AAM. Intestinal cell migration damage induced by enteropathogenic Escherichia coli strains. ACTA ACUST UNITED AC 2018; 51:e7423. [PMID: 30066727 PMCID: PMC6065879 DOI: 10.1590/1414-431x20187423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/24/2018] [Indexed: 01/27/2023]
Abstract
Epithelial cell migration is an essential response to enteric pathogens such as enteropathogenic Escherichia coli (EPEC). This study aimed to investigate the effects of EPEC infection on intestinal epithelial cell migration in vitro, as well as the involvement of type III secretion system (T3SS) and Rho GTPases. Crypt intestinal epithelial cells (IEC-6) were infected with EPEC strains (E2348/69, ΔescF, and the LDI001 strain isolated from a malnourished Brazilian child) and commensal E. coli HS. Wound migration and cell death assays were performed at different time-points. Transcription and expression of Rho GTPases were evaluated using real-time PCR and western blotting. Overall, EPEC E2348/69 reduced migration and increased apoptosis and necrosis levels compared to EPEC LDI001 and E. coli HS strains. Moreover, EPEC LDI001 impaired cell migration at a higher level than E. coli HS and increased necrosis after 24 hours compared to the control group. The different profiles of virulence genes between the two wild-type EPEC strains, characterized by the absence of espL and nleE genes in the LDI001, might explain the phenotypic results, playing significant roles on cell migration impairment and cell death-related events. Moreover, the type III secretion system is determinant for the inhibition of intestinal epithelial cell migration by EPEC 2348/69, as its deletion prevented the effect. Active Rac1 concentrations were increased in E2348/69 and LDI001-infected cells, while the T3SS-deficient strain did not demonstrate this activation. This study contributes with valuable insight to characterize the mechanisms involved in the impairment of intestinal cell migration induced by EPEC.
Collapse
Affiliation(s)
- P A Cavalcante
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M M G Prata
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P H Q S Medeiros
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A V Alves da Silva
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - J S Quetz
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M A V Reyes
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T S Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A K S Santos
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S A Ribeiro
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - H N Veras
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M D Bona
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M S M G Amaral
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - F A P Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - I F N Lima
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A Havt
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A A M Lima
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
7
|
Liu Z, Huang C, Liu Y, Lin D, Zhao Y. NMR-based metabolomic analysis of the effects of alanyl-glutamine supplementation on C2C12 myoblasts injured by energy deprivation. RSC Adv 2018; 8:16114-16125. [PMID: 35542200 PMCID: PMC9080260 DOI: 10.1039/c8ra00819a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022] Open
Abstract
The dipeptide alanyl-glutamine (Ala-Gln) is a well-known parenteral nutritional supplement. The Ala-Gln supplementation is a potential treatment for muscle-related diseases and injuries. However, molecular mechanisms underlying the polyphenic effects of Ala-Gln supplementation remain elusive. Here, we performed NMR-based metabolomic profiling to analyze the effects of Ala-Gln, and the free alanine (Ala) and glutamine (Gln) supplementations on the mouse myoblast cell line C2C12 injured by glucose and glutamine deprivation. All the three supplementations can promote the differentiation ability of the injured C2C12 cells, while only Ala-Gln supplementation can facilitate the proliferation of the injured cells. Ala-Gln supplementation can partially restore the metabolic profile of C2C12 myoblasts disturbed by glucose and glutamine deprivation, and exhibits more significant effects than Ala and Gln supplementations. Our results suggest that Ala-Gln supplementation can promote MyoD1 protein synthesis, upregulate the muscle ATP-storage phosphocreatine (PCr), maintain TCA cycle anaplerosis, enhance the antioxidant capacity through promoting GSH biosynthesis, and stabilize lipid membranes by suppressing glycerophospholipids metabolism. This work provides new insight into mechanistic understanding of the polyphenic effects of Ala-Gln supplementation on muscle cells injured by energy deprivation.
Collapse
Affiliation(s)
- Zhiqing Liu
- College of Chemistry and Chemical Engineering, The Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Xiamen University Xiamen 361005 China +86-592-218-6078 +86-592-218-5610
| | - Caihua Huang
- Exercise and Health Laboratory, Xiamen University of Technology Xiamen 361024 China
| | - Yan Liu
- College of Chemistry and Chemical Engineering, The Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Xiamen University Xiamen 361005 China +86-592-218-6078 +86-592-218-5610
| | - Donghai Lin
- College of Chemistry and Chemical Engineering, The Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Xiamen University Xiamen 361005 China +86-592-218-6078 +86-592-218-5610
| | - Yufen Zhao
- College of Chemistry and Chemical Engineering, The Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Xiamen University Xiamen 361005 China +86-592-218-6078 +86-592-218-5610
| |
Collapse
|
8
|
Xing S, Zhang B, Lin M, Zhou P, Li J, Zhang L, Gao F, Zhou G. Effects of alanyl-glutamine supplementation on the small intestinal mucosa barrier in weaned piglets. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 30:236-245. [PMID: 27383799 PMCID: PMC5205612 DOI: 10.5713/ajas.16.0077] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/27/2016] [Accepted: 06/28/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The study was to investigate the effects of alanyl-glutamine (Ala-Gln) and glutamine (Gln) supplementation on the intestinal mucosa barrier in piglets. METHODS A total of 180 barrows with initial weight 10.01±0.03 kg were randomly allocated to three treatments, and each treatment consisted of three pens and twenty pigs per pen. The piglets of three groups were fed with control diet [0.62% alanine (Ala)], Ala-Gln diet (0.5% Ala-Gln), Gln diet (0.34% Gln and 0.21% Ala), respectively. RESULTS The results showed that in comparison with control diet, dietary Ala-Gln supplementation increased the height of villi in duodenum and jejunum (p<0.05), Gln supplementation increased the villi height of jejunum (p<0.05), Ala-Gln supplementation up-regulated the mRNA expressions of epidermal growth factor receptor and insulin-like growth factor 1 receptor in jejunal mucosa (p<0.05), raised the mRNA expressions of Claudin-1, Occludin, zonula occludens protein-1 (ZO-1) and the protein levels of Occludin, ZO-1 in jejunal mucosa (p<0.05), Ala-Gln supplementation enlarged the number of goblet cells in duodenal and ileal epithelium (p<0.05), Gln increased the number of goblet cells in duodenal epithelium (p<0.05) and Ala-Gln supplementation improved the concentrations of secretory immunoglobulin A and immunoglobulin G in the jejunal mucosa (p<0.05). CONCLUSION These results demonstrated that dietary Ala-Gln supplementation could maintain the integrity of small intestine and promote the functions of intestinal mucosa barriers in piglets.
Collapse
Affiliation(s)
- Shen Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Bolin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China.,Department of Agricultural Science and Technology, Zunyi Normal College, Zunyi 563002, China
| | - Meng Lin
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Zhou
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiaolong Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Guanghong Zhou
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
9
|
Venoji R, Amirtharaj GJ, Kini A, Vanaparthi S, Venkatraman A, Ramachandran A. Enteral glutamine differentially regulates Nrf 2 along the villus-crypt axis of the intestine to enhance glutathione levels. J Gastroenterol Hepatol 2015; 30:1740-7. [PMID: 26095579 DOI: 10.1111/jgh.13019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/23/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Glutamine is an important energy source for the intestinal epithelium, and its supplementation protects intestinal epithelial cells by induction of glutathione. However, mechanisms of glutathione induction in cells at various stages of differentiation along the crypt to villus axis are not well understood. This study examined induction of glutathione in response to glutamine along the intestinal villus-crypt axis and evaluated regulatory mediators involved in the process. METHODS Animals were administered 4% glutamine in feed for 7 days, following which enterocytes at various stages of differentiation were isolated and glutathione levels and signaling mediators involved in its regulation were studied. RESULTS In control animals, glutathione levels were higher in the intestinal crypt than in the villus or middle region. This was accompanied by elevated expression of the modifier subunit of glutathione synthetase (GCLM) and the transcription factor Nrf2 when compared with cells from the villus and middle regions. These levels were further enhanced by glutamine throughout the intestine, although the effects were more dramatic in the crypt. In parallel to glutathione induction, glutamine supplementation also altered actin dynamics and proliferation in cells of the crypt. CONCLUSIONS These results suggest that the variation of glutathione levels along the villus-crypt axis in the intestine is due to gradients in expression of mediators such as glutamate cysteine ligase modifier subunit and Nrf2. The protective effects of glutamine supplementation seem to be most pronounced in the crypt, where it upregulates proliferation, glutathione levels and alters actin dynamics.
Collapse
Affiliation(s)
- Raghupathy Venoji
- Wellcome Trust Research Laboratory, Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Gnanaraj Jayakumar Amirtharaj
- Wellcome Trust Research Laboratory, Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Archana Kini
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Sivakumar Vanaparthi
- Wellcome Trust Research Laboratory, Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Aparna Venkatraman
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Anup Ramachandran
- Wellcome Trust Research Laboratory, Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| |
Collapse
|
10
|
Araújo CV, Lazzarotto CR, Aquino CC, Figueiredo IL, Costa TB, Alves LADO, Ribeiro RA, Bertolini LR, Lima AAM, Brito GAC, Oriá RB. Alanyl-glutamine attenuates 5-fluorouracil-induced intestinal mucositis in apolipoprotein E-deficient mice. ACTA ACUST UNITED AC 2015; 48:493-501. [PMID: 25945744 PMCID: PMC4470307 DOI: 10.1590/1414-431x20144360] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/11/2014] [Indexed: 01/19/2023]
Abstract
Apolipoprotein E (APOE=gene, apoE=protein) is a known factor regulating the inflammatory response that may have regenerative effects during tissue recovery from injury. We investigated whether apoE deficiency reduces the healing effect of alanyl-glutamine (Ala-Gln) treatment, a recognized gut-trophic nutrient, during tissue recovery after 5-FU-induced intestinal mucositis. APOE-knockout (APOE-/-) and wild-type (APOE+/+) C57BL6J male and female mice (N=86) were given either Ala-Gln (100 mM) or phosphate buffered saline (PBS) by gavage 3 days before and 5 days after a 5-fluorouracil (5-FU) challenge (450 mg/kg, via intraperitoneal injection). Mouse body weight was monitored daily. The 5-FU cytotoxic effect was evaluated by leukometry. Intestinal villus height, villus/crypt ratio, and villin expression were monitored to assess recovery of the intestinal absorptive surface area. Crypt length, mitotic, apoptotic, and necrotic crypt indexes, and quantitative real-time PCR for insulin-like growth factor-1 (IGF-1) and B-cell lymphoma 2 (Bcl-2) intestinal mRNA transcripts were used to evaluate intestinal epithelial cell turnover. 5-FU challenge caused significant weight loss and leukopenia (P<0.001) in both mouse strains, which was not improved by Ala-Gln. Villus blunting, crypt hyperplasia, and reduced villus/crypt ratio (P<0.05) were found in all 5-FU-challenged mice but not in PBS controls. Ala-Gln improved villus/crypt ratio, crypt length and mitotic index in all challenged mice, compared with PBS controls. Ala-Gln improved villus height only in APOE-/- mice. Crypt cell apoptosis and necrotic scores were increased in all mice challenged by 5-FU, compared with untreated controls. Those scores were significantly lower in Ala-Gln-treated APOE+/+ mice than in controls. Bcl-2 and IGF-1 mRNA transcripts were reduced only in the APOE-/- -challenged mice. Altogether our findings suggest APOE-independent Ala-Gln regenerative effects after 5-FU challenge.
Collapse
Affiliation(s)
- C V Araújo
- Laboratório da Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - C R Lazzarotto
- Laboratório de Biologia Molecular e do Desenvolvimento, Universidade de Fortaleza, Fortaleza, CE, Brasil
| | - C C Aquino
- Laboratório da Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - I L Figueiredo
- Laboratório da Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T B Costa
- Laboratório da Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - L A de Oliveira Alves
- Laboratório da Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R A Ribeiro
- Laboratório da Inflamação e Câncer, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - L R Bertolini
- Laboratório de Biologia Molecular e do Desenvolvimento, Universidade de Fortaleza, Fortaleza, CE, Brasil
| | - A A M Lima
- Laboratório de Doenças Infecciosas, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - G A C Brito
- Laboratório da Inflamação e Câncer, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R B Oriá
- Laboratório da Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, INCT - Instituto de Biomedicina do Semiárido Brasileiro, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
11
|
Hu K, Feng L, Jiang W, Liu Y, Jiang J, Li S, Zhou X. Oxidative damage repair by glutamine in fish enterocytes. FISH PHYSIOLOGY AND BIOCHEMISTRY 2014; 40:1437-1445. [PMID: 24777750 DOI: 10.1007/s10695-014-9938-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/04/2014] [Indexed: 06/03/2023]
Abstract
Fish intestine is very sensitive to oxidative damage. Repair of damaged enterocytes may be involved to restore normal function of fish intestine. However, studies of fish enterocyte repair are scarce. The present study aimed to investigate the potential repair role of glutamine after a H2O2 challenge. In this study, fish enterocytes were post-treated with graded levels of glutamine (0, 4, 8, 12 and 20 mM of glutamine) after expose to 100 μM H2O2. The basal control cells were kept in the glutamine-free minimum essential medium only. Results showed that the H2O2-induced decreases in 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide optical density, alkaline phosphatase and Na(+), K(+)-ATPase activities were completely restored by subsequent glutamine treatments. In addition, cellular injury (lactate dehydrogenase), lipid peroxidation (malondialdehyde) and protein oxidation (protein carbonyls) caused by H2O2 were reversed by subsequent glutamine treatments. Furthermore, the H2O2-induced decreases in glutathione contents, glutathione reductase, superoxide dismutase and glutathione peroxidase activities were completely restored by subsequent glutamine treatments. In summary, the present study indicated that glutamine improved the repair activity in fish enterocytes after challenge with H2O2.
Collapse
Affiliation(s)
- Kai Hu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Swaid F, Sukhotnik I, Matter I, Berkowitz D, Hadjittofi C, Pollak Y, Lavy A. Dietary glutamine supplementation prevents mucosal injury and modulates intestinal epithelial restitution following acetic acid induced intestinal injury in rats. Nutr Metab (Lond) 2013; 10:53. [PMID: 23919638 PMCID: PMC3750704 DOI: 10.1186/1743-7075-10-53] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 07/23/2013] [Indexed: 12/27/2022] Open
Abstract
Beneficial effects of glutamine (GLN) have been described in many gastrointestinal disorders. The aim of the present study was to evaluate the preventative effect of oral GLN supplementation against acetic acid (AA) induced intestinal injury in a rat. Male Sprague-Dawley rats were divided into four experimental groups: control (CONTR) rats underwent laparotomy, control-glutamine (CONTR-GLN) rats were treated with enteral glutamine given in drinking water (2%) 48 hours before and five days following laparotomy, AA rats underwent laparotomy and injection of AA into an isolated jejunal loop, and acetic acid-glutamine (AA-GLN) rats underwent AA-induced injury and were treated with enteral GLN 48 hours before and 5 days following laparotomy. Intestinal mucosal damage (Park's injury score), mucosal structural changes, enterocyte proliferation and enterocyte apoptosis were determined five days following intestinal injury. Western blotting was used to determine p-ERK and bax protein levels. AA-induced intestinal injury resulted in a significantly increased intestinal injury score with concomitant inhibition of cell turnover (reduced proliferation and enhanced apoptosis). Treatment with dietary GLN supplementation resulted in a decreased intestinal injury score with concomitant stimulation of cell turnover (enhanced proliferation and reduced apoptosis). In conclusion, pre-treatment with oral GLN prevents mucosal injury and improves intestinal recovery following AA-induced intestinal injury in rats.
Collapse
Affiliation(s)
- Forat Swaid
- Department of Surgery, Bnai Zion Medical Center, Haifa, Israel
| | - Igor Sukhotnik
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department Pediatric Surgery, Bnai Zion Medical Center, 47 Golomb St., POB 4940, Haifa 31048, Israel
| | - Ibrahim Matter
- Department of Surgery, Bnai Zion Medical Center, Haifa, Israel
| | - Drora Berkowitz
- Department of Gastroenterology, Bnai Zion Medical Center, Haifa, Israel
| | - Christopher Hadjittofi
- Elderly Care Department, Queen Elizabeth II Hospital, Welwyn Garden City, United Kingdom
| | - Yulia Pollak
- Laboratory of Intestinal Adaptation and Recovery, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Alexandra Lavy
- Department of Gastroenterology, Bnai Zion Medical Center, Haifa, Israel
| |
Collapse
|
13
|
Rodrigues RS, Oliveira RAC, Li Y, Zaja-Milatovic S, Costa LB, Braga Neto MB, Kolling GL, Lima AA, Guerrant RL, Warren CA. Intestinal epithelial restitution after TcdB challenge and recovery from Clostridium difficile infection in mice with alanyl-glutamine treatment. J Infect Dis 2013; 207:1505-15. [PMID: 23359592 DOI: 10.1093/infdis/jit041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Clostridium difficile is an anaerobic bacterium that causes antibiotic-associated diarrhea. It produces toxin A and toxin B (TcdB), which cause injury to the gut epithelium. Glutamine is a fundamental fuel for enterocytes, maintaining intestinal mucosal health. Alanyl-glutamine (AQ) is a highly soluble dipeptide derivative of glutamine. We studied whether administration of AQ ameliorates the effects of TcdB in the intestinal cells and improves the outcome of C. difficile infection in mice. METHODS WST-1 proliferation and cell-wounding-migration assays were assessed in IEC-6 cells exposed to TcdB, with or without AQ. Apoptosis and necrosis were assessed using Annexin V and flow cytometry. C57BL/6 mice were infected with VPI 10463 and treated with either vancomycin, AQ, or vancomycin with AQ. Intestinal tissues were collected for histopathologic analysis, apoptosis staining, and determination of myeloperoxidase activity. RESULTS AQ increased proliferation in intestinal cells exposed to TcdB, improved migration at 24 and 48 hours, and reduced apoptosis in intestinal cells challenged with TcdB. Infected mice treated with vancomycin and AQ had better survival and histopathologic findings than mice treated with vancomycin alone. CONCLUSIONS AQ may reduce intestinal mucosal injury in C. difficile-infected mice by partially reversing the effects of TcdB on enterocyte proliferation, migration, and apoptosis, thereby improving survival from C. difficile infection.
Collapse
|
14
|
Glutamine and alanyl-glutamine increase RhoA expression and reduce Clostridium difficile toxin-a-induced intestinal epithelial cell damage. BIOMED RESEARCH INTERNATIONAL 2012; 2013:152052. [PMID: 23484083 PMCID: PMC3591182 DOI: 10.1155/2013/152052] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/12/2012] [Indexed: 12/18/2022]
Abstract
Clostridium difficile is a major cause of antibiotic-associated colitis and is associated with significant morbidity and mortality. Glutamine (Gln) is a major fuel for the intestinal cell population. Alanyl-glutamine (Ala-Gln) is a dipeptide that is highly soluble and well tolerated. IEC-6 cells were used in the in vitro experiments. Cell morphology was evaluated by atomic force microscopy (AFM) and scanning electron microscopy (SEM). Cell proliferation was assessed by WST-1 and Ki-67 and apoptosis was assessed by TUNEL. Cytoskeleton was evaluated by immunofluorescence for RhoA and F-actin. RhoA was quantified by immunoblotting. TcdA induced cell shrinkage as observed by AFM, SEM, and fluorescent microscopy. Additionally, collapse of the F-actin cytoskeleton was demonstrated by immunofluorescence. TcdA decreased cell volume and area and increased cell height by 79%, 66.2%, and 58.9%, respectively. Following TcdA treatment, Ala-Gln and Gln supplementation, significantly increased RhoA by 65.5% and 89.7%, respectively at 24 h. Ala-Gln supplementation increased cell proliferation by 137.5% at 24 h and decreased cell apoptosis by 61.4% at 24 h following TcdA treatment. In conclusion, TcdA altered intestinal cell morphology and cytoskeleton organization, decreased cell proliferation, and increased cell apoptosis. Ala-Gln and Gln supplementation reduced intestinal epithelial cell damage and increased RhoA expression.
Collapse
|
15
|
Braga-Neto MB, Oliveira BMC, Rodrigues RS, Noronha FJ, Leitao RF, Brito GAC, Lima AA, Guerrant RL, Warren CA. Protective effects of alanyl-glutamine supplementation against nelfinavir-induced epithelial impairment in IEC-6 cells and in mouse intestinal mucosa. Cancer Biol Ther 2012; 13:1482-90. [PMID: 22986234 DOI: 10.4161/cbt.22251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Human Immunodeficiency Virus (HIV) protease inhibitors (PI) remain a crucial component of highly active therapy (HAART) and recently have been demonstrated to have potent antitumor effect on a wide variety of tumor cell lines. However, discontinuation of therapy is an important issue, which may be related to various side-effects, especially diarrhea. The aim of this study was to evaluate the effects of nelfinavir (NFV), an HIV PI, and of alanyl-glutamine (AQ) supplementation, on intestinal cell migration, proliferation, apoptosis and necrosis, using IEC-6 cells and on intestinal crypt depth, villus length, villus area, mitotic index and apoptosis in Swiss mice. METHODS Migration was evaluated at 12 and 24 h after injury using a wound healing assay. Cellular proliferation was measured indirectly at 24 and 48 h using tetrazolium salt WST-1. Apoptosis and necrosis were measured by flow cytometry using the Annexin V assay. Intestinal morphometry and mitotic index in vivo were assessed following a seven-day treatment with 100 mg/kg of NFV, given orally. In vivo proliferation and apoptosis were evaluated by intestinal crypt mitotic index and immunohistochemistry, respectively. RESULTS In vitro, AQ supplementation enhanced IEC-6 cell migration and proliferation, following challenge with NFV. In vivo, AQ increased intestinal villus length, villus area, crypt depth and cell proliferation and cell migration, following treatment with NFV. AQ did not decrease cell death induced by NFV both in vivo and in vitro. CONCLUSIONS AQ supplementation is potentially beneficial in preventing the effects of PIs, such as NFV, in the intestinal tract.
Collapse
Affiliation(s)
- Manuel B Braga-Neto
- Clinical Research Unit & Institute of Biomedicine/Center for Global Health, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Azevedo OGR, Oliveira RAC, Oliveira BC, Zaja-Milatovic S, Araújo CV, Wong DVT, Costa TB, Lucena HBM, Lima RCP, Ribeiro RA, Warren CA, Lima AÂM, Vitek MP, Guerrant RL, Oriá RB. Apolipoprotein E COG 133 mimetic peptide improves 5-fluorouracil-induced intestinal mucositis. BMC Gastroenterol 2012; 12:35. [PMID: 22524518 PMCID: PMC3398852 DOI: 10.1186/1471-230x-12-35] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 04/23/2012] [Indexed: 12/27/2022] Open
Abstract
Background Intestinal mucositis is one of the major troublesome side effects of anticancer chemotherapy leading to poor patient compliance. In this study we addressed the role of the novel apolipoprotein E (ApoE) COG 133 mimetic peptide in 5-fluorouracil (5-FU)-challenged Swiss mice and IEC-6 cell monolayers. Experiments were also conducted in C57BL6J ApoE knock-out mice to assess the effects of apoE peptide treatment. Methods Experimental groups were as follows: unchallenged controls, 5-FU-challenged mice (450 mg/kg, i.p) with or without the ApoE peptide (0.3, 1, and 3 μM, given twice daily i.p. for 4 days). Mice were sacrificed 3 days after 5-FU challenge. Proximal small intestinal samples were harvested for molecular biology and histological processing. We conducted ELISA assays and RT-PCR to target IL-1β, TNF-α, IL-10, iNOS, and myeloperoxidase (MPO) to assess intestinal inflammation. Cell death and NF-κB assays were also conducted in apoE knock-out mice. In our in vitro models, IEC-6 cells were exposed to 1 mM of 5-FU in glutamine free media with or without the ApoE peptide (0.02, 0.2, 2, 5, 10, and 20 μM). We investigated IEC-6 cell proliferation and migration, 24 h after the 5-FU challenge. Additionally, apoptotic IEC-6 cells were measured by Tunel and flow cytometry. Equimolar doses of the ApoA-I (D4-F) peptide were also used in some experiments for comparative studies. Results Villus blunting and heavy inflammatory infiltrates were seen in the 5-FU-challenged group, findings that were partially ameliorated by the ApoE peptide. We found increased intestinal MPO and pro-inflammatory IL-1β and TNF-α levels, and TNF-α and iNOS transcripts, and reduction of IL-10 following 5-FU treatment, each of which were partially abrogated by the peptide. Improvements were also found in IEC-6 cell apoptosis and migration following ApoE and D-4F treatment. Conclusion Altogether, these findings suggest that the novel ApoE COG 133 mimetic peptide can reduce 5-FU-induced intestinal changes and potentially benefit mucositis.
Collapse
Affiliation(s)
- Orleâncio Gomes R Azevedo
- Center for Global Health, School of Medicine, University of Virginia, Carter Harrison Bldg MR-6, 625 Crispell Drive, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Costa LB, Noronha FJ, Roche JK, Sevilleja JE, Warren CA, Oriá R, Lima A, Guerrant RL. Novel in vitro and in vivo models and potential new therapeutics to break the vicious cycle of Cryptosporidium infection and malnutrition. J Infect Dis 2012; 205:1464-71. [PMID: 22454464 DOI: 10.1093/infdis/jis216] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although several animal models of cryptosporidiosis have been reported, most involve genetically or pharmacologically immune-suppressed hosts. METHODS We report challenge with excysted (in vitro and in vivo) and unexcysted (in vivo) Cryptosporidium parvum oocysts in human colonic adenocarcinoma (HCT-8) cells and weaned nourished and malnourished C57BL/6 mice, following outcomes of growth rate, stool shedding, and tissue burden. We tested treatment with an oligodeoxynucleotide containing unmethylated CpG motif (CpG-ODN) and alanyl-glutamine in vivo and in vitro. RESULTS C. parvum-challenged mice showed prolonged weight loss (>10% over 4 days), robust stool shedding (>3 logs/d over 7 days), and epithelial infection in the ileum, cecum, and colon. Of 2 potential therapeutic compounds evaluated in the model, CpG-ODN reduced body weight loss (to <6% on days 3-7 after challenge), reduced shedding of organisms (by 25% on days 1 and 3 after challenge), and decreased the burden of parasites in the ileum. Alanyl-glutamine showed similar benefits. In vitro findings suggested that effects on the epithelial component of the mucosa probably likely responsible for beneficial effects seen in vivo. CONCLUSIONS Weaned mice provide a convenient and reproducible model of cryptosporidial disease, including its vicious cycle with body weight loss and heavier infection with malnutrition, and this model may be useful in exploring innovative therapeutic solutions for this challenging infectious disease.
Collapse
Affiliation(s)
- Lourrany B Costa
- Center for Global Health, Division of Infectious Diseases and International Health, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Warren CA, Calabrese GM, Li Y, Pawlowski SW, Figler RA, Rieger J, Ernst PB, Linden J, Guerrant RL. Effects of adenosine A₂A receptor activation and alanyl-glutamine in Clostridium difficile toxin-induced ileitis in rabbits and cecitis in mice. BMC Infect Dis 2012; 12:13. [PMID: 22264229 PMCID: PMC3323464 DOI: 10.1186/1471-2334-12-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 01/20/2012] [Indexed: 12/18/2022] Open
Abstract
Background Severe Clostridium difficile toxin-induced enteritis is characterized by exuberant intestinal tissue inflammation, epithelial disruption and diarrhea. Adenosine, through its action on the adenosine A2A receptor, prevents neutrophillic adhesion and oxidative burst and inhibits inflammatory cytokine production. Alanyl-glutamine enhances intestinal mucosal repair and decreases apoptosis of enterocytes. This study investigates the protection from enteritis by combination therapy with ATL 370, an adenosine A2A receptor agonist, and alanyl-glutamine in a rabbit and murine intestinal loop models of C. difficile toxin A-induced epithelial injury. Methods Toxin A with or without alanyl-glutamine was administered intraluminally to rabbit ileal or murine cecal loops. Animals were also given either PBS or ATL 370 parenterally. Ileal tissues were examined for secretion, histopathology, apoptosis, Cxcl1/KC and IL-10. Results ATL 370 decreased ileal secretion and histopathologic changes in loops treated with Toxin A. These effects were reversed by the A2A receptor antagonist, SCH 58261, in a dose-dependent manner. The combination of ATL 370 and alanyl-glutamine significantly further decreased ileal secretion, mucosal injury and apoptosis more than loops treated with either drug alone. ATL 370 and alanyl-glutamine also decreased intestinal tissue KC and IL-10. Conclusions Combination therapy with an adenosine A2A receptor agonist and alanyl-glutamine is effective in reversing C. difficile toxin A-induced epithelial injury, inflammation, secretion and apoptosis in animals and has therapeutic potential for the management of C. difficile infection.
Collapse
Affiliation(s)
- Cirle Alcantara Warren
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Down-regulation of glutamine synthetase enhances migration of rat astrocytes after in vitro injury. Neurochem Int 2010; 58:404-13. [PMID: 21193003 DOI: 10.1016/j.neuint.2010.12.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 12/20/2010] [Accepted: 12/20/2010] [Indexed: 11/23/2022]
Abstract
Astrocytes undergo reactive transformation in response to physical injury (reactive gliosis) that may impede neural repair. Glutamine synthetase (GS) is highly expressed by astrocytes, and serves a neuroprotective function by converting cytotoxic glutamate and ammonia into glutamine. Glutamine synthetase was down-regulated in reactive astrocytes at the site of mechanical spinal cord injury (SCI) and in cultured astrocytes at the margins of a scratch wound, suggesting that GS may modulate reactive transformation and glial scar development. We evaluated this potential function of GS using siRNA-mediated GS knock-down. Suppression of astrocytic GS by GS siRNA increased cell migration into the scratch wound zone and decreased substrate adhesion as indicated by the number of focal adhesions expressing the adaptor protein paxillin. Migration was enhanced by glutamine and suppressed by glutamate, in contrast to the result expected if enhanced migration was due solely to changes in glutamine and glutamate concomitant with reduced GS activity. The membrane type 1-matrix metalloproteinase (MT1-MMP) was up-regulated in GS siRNA-treated astrocytes, while a broad-spectrum MMP antagonist inhibited migration in both wild type and GS knock-down astrocytes. In addition, GS siRNA inhibited expression of integrin β1, while antibody-mediated inhibition of integrin β1 impaired direction-specific protrusion and motility. Thus, GS may modulate motility and substrate adhesion through transmembrane integrin β1 signaling to the cytoskeleton and by MMT-mediated proteolysis of the extracellular matrix.
Collapse
|
20
|
Boukhettala N, Ibrahim A, Claeyssens S, Faure M, Le Pessot F, Vuichoud J, Lavoinne A, Breuillé D, Déchelotte P, Coëffier M. A diet containing whey protein, glutamine, and TGFbeta modulates gut protein metabolism during chemotherapy-induced mucositis in rats. Dig Dis Sci 2010; 55:2172-81. [PMID: 19911274 DOI: 10.1007/s10620-009-1039-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Accepted: 10/26/2009] [Indexed: 02/01/2023]
Abstract
BACKGROUND Mucositis, a common side effect of chemotherapy, is characterized by compromised digestive function, barrier integrity and immune competence. AIMS Our aim was to evaluate the impact of a specifically designed diet Clinutren Protect (CP), which contains whey proteins, TGFbeta-rich casein, and free glutamine, on mucositis in rats. METHODS Mucositis was induced by three consecutive injections (day 0, day 1, day 2) of methotrexate (2.5 mg/kg). Rats had free access to CP or placebo diets from days -7 to 9. In the placebo diet, whey proteins and TGFbeta-rich casein were replaced by TGFbeta-free casein and glutamine by alanine. Intestinal parameters were assessed at day 3 and 9. Values, expressed as mean +/- SEM, were compared using two-way ANOVA. RESULTS At day 3, villus height was markedly decreased in the placebo (296 +/- 11 microm) and CP groups (360 +/- 10 microm) compared with controls (464 +/- 27 microm), but more markedly in the placebo as compared to CP group. The intestinal damage score was also reduced in the CP compared with the placebo group. Glutathione content increased in the CP compared with the placebo group (2.2 +/- 0.2 vs. 1.7 +/- 0.2 micromol/g tissue). Gut protein metabolism was more affected in the placebo than in the CP group. The fractional synthesis rate was decreased in the placebo group (93.8 +/- 4.9%/day) compared with controls (121.5 +/- 12.1, P < 0.05), but not in the CP group (106.0 +/- 13.1). In addition, at day 9, rats exhibited improved body weight and food intake recovery in the CP compared to the placebo group. CONCLUSIONS Clinutren Protect feeding reduces intestinal injury in the acute phase of methotrexate-induced mucositis in rats and improves recovery.
Collapse
Affiliation(s)
- Nabile Boukhettala
- ADEN EA4311 Institute for Biomedical Research and European Institute for Peptide Research (IFRMP23), Rouen University, Rouen, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Preidis GA, Versalovic J. Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology 2009; 136:2015-31. [PMID: 19462507 PMCID: PMC4108289 DOI: 10.1053/j.gastro.2009.01.072] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Studies of metagenomics and the human microbiome will tremendously expand our knowledge of the composition of microbial communities in the human body. As our understanding of microbial variation and corresponding genetic parameters is refined, this information can be applied to rational remodeling or "tailoring" of human-associated microbial communities and their associated functions. Physiologic features such as the development of innate and adaptive immunity, relative susceptibilities to infections, immune tolerance, bioavailability of nutrients, and intestinal barrier function may be modified by changing the composition and functions of the microbial communities. The specialty of gastroenterology will be affected profoundly by the ability to modify the gastrointestinal microbiota through the rational deployment of antibiotics, probiotics, and prebiotics. Antibiotics might be used to remove or suppress undesirable components of the human microbiome. Probiotics can introduce missing microbial components with known beneficial functions for the human host. Prebiotics can enhance the proliferation of beneficial microbes or probiotics, to maximize sustainable changes in the human microbiome. Combinations of these approaches might provide synergistic and effective therapies for specific disorders. The human microbiome could be manipulated by such "smart" strategies to prevent and treat acute gastroenteritis, antibiotic-associated diarrhea and colitis, inflammatory bowel disease, irritable bowel syndrome, necrotizing enterocolitis, and a variety of other disorders.
Collapse
Affiliation(s)
- Geoffrey A. Preidis
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
- Departments of Pathology, Texas Children’s Hospital and Baylor College of Medicine, Houston, Texas
| | - James Versalovic
- Departments of Pathology, Texas Children’s Hospital and Baylor College of Medicine, Houston, Texas
| |
Collapse
|