1
|
Nazir U, Fu Z, Zheng X, Zafar MH, Chen Y, Yang Z, Wang Z, Yang H. Effects of Alanyl-Glutamine Dipeptide Supplementation on Growth Performance, Nutrient Digestibility, Digestive Enzyme Activity, Immunity, and Antioxidant Status in Growing Laying Hens. Animals (Basel) 2024; 14:2934. [PMID: 39457865 PMCID: PMC11503830 DOI: 10.3390/ani14202934] [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: 09/04/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Alanyl-glutamine (Aln-Gln), a highly soluble and stable Glutamine-dipeptide, is known to improve the performance of poultry birds. This study aimed to investigate the effect of Aln-Gln during the rearing period on growth performance, nutrient digestibility, digestive enzyme activity, immunity, antioxidant status and relative gene expression of Hy-Line brown hens. A total of 480 healthy day-old Hy-line brown chicks with similar body weights were randomly divided into four dietary groups (8 replicates/group and 15 birds/replicate). Groups A, B, C and D were fed diets containing 0%, 0.1%, 0.2% and 0.3% Aln-Gln, respectively, for 6 weeks. The body weight (BW) and average daily gain (ADG) were higher in hens fed test diets compared with the control (p < 0.05). The feed conversion ratio (FCR) was better in test groups as compared to the control group (p < 0.05). The ADFI showed no significant difference between the groups. Dietary treatments had no effect on dry matter (DM), organic matter (OM) and crude fiber (CF) digestibility. The Aln-Gln also improved gross energy (GE) and crude protein (CP) digestibility (p < 0.05). It has also increased IgG levels in groups C and D. IgM levels were similar to the control in B, C and D. The Aln-Gln increased IL-1 in B and C, IL-2 in C and D, and IL-6 in all test groups (p < 0.05). The supplementation of Aln-Gln had no effect on serum antioxidant indices like CAT, MDA, GSH-PX, GSH, and SOD in 42-day-old growing hens. Aln-Gln supplementation had no significant effect (p > 0.05) on the activity of amylase and lipase, however, a significant improvement (p < 0.05) in the activities of trypsin and chymotrypsin was observed in the test groups. Supplemented Aln-Gln levels in the birds' diets led to an increase in the expression of genes related to growth factors (IGF-1, IGFBP-5), immune markers (IL-1, IL-2, IL-6) and antioxidant status (GSH-Px1), as compared to control group. Aln-Gln supplementation in Hy-Line brown hens during their growing period improved growth, nutrient digestibility, immunity and digestive enzymes activity. These findings suggest that Aln-Gln is a promising dietary additive for enhancing poultry performance.
Collapse
Affiliation(s)
- Usman Nazir
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhenming Fu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xucheng Zheng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Muhammad Hammad Zafar
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yuanjing Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhi Yang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Zhiyue Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Haiming Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
2
|
Ran Z, Yang J, Liu L, Wu S, An Y, Hou W, Cheng T, Zhang Y, Zhang Y, Huang Y, Zhang Q, Wan J, Li X, Xing B, Ye Y, Xu P, Chen Z, Zhao J, Li R. Chronic PM 2.5 exposure disrupts intestinal barrier integrity via microbial dysbiosis-triggered TLR2/5-MyD88-NLRP3 inflammasome activation. ENVIRONMENTAL RESEARCH 2024; 258:119415. [PMID: 38906446 DOI: 10.1016/j.envres.2024.119415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND PM2.5, a known public health risk, is increasingly linked to intestinal disorders, however, the mechanisms of its impact are not fully understood. PURPOSE This study aimed to explore the impact of chronic PM2.5 exposure on intestinal barrier integrity and to uncover the underlying molecular mechanisms. METHODS C57BL/6 J mice were exposed to either concentrated ambient PM2.5 (CPM) or filtered air (FA) for six months to simulate urban pollution conditions. We evaluated intestinal barrier damage, microbial shifts, and metabolic changes through histopathology, metagenomics, and metabolomics. Analysis of the TLR signaling pathway was also conducted. RESULTS The mean concentration of PM2.5 in the CPM exposure chamber was consistently measured at 70.9 ± 26.8 μg/m³ throughout the study period. Our findings show that chronic CPM exposure significantly compromises intestinal barrier integrity, as indicated by reduced expression of the key tight junction proteins Occludin and Tjp1/Zo-1. Metagenomic sequencing revealed significant shifts in the microbial landscape, identifying 35 differentially abundant species. Notably, there was an increase in pro-inflammatory nongastric Helicobacter species and a decrease in beneficial bacteria, such as Lactobacillus intestinalis, Lactobacillus sp. ASF360, and Eubacterium rectale. Metabolomic analysis further identified 26 significantly altered metabolites commonly associated with intestinal diseases. A strong correlation between altered bacterial species and metabolites was also observed. For example, 4 Helicobacter species all showed positive correlations with 13 metabolites, including Lactate, Bile acids, Pyruvate and Glutamate. Additionally, increased expression levels of TLR2, TLR5, Myd88, and NLRP3 proteins were noted, and their expression patterns showed a strong correlation, suggesting a possible involvement of the TLR2/5-MyD88-NLRP3 signaling pathway. CONCLUSIONS Chronic CPM exposure induces intestinal barrier dysfunction, microbial dysbiosis, metabolic imbalance, and activation of the TLR2/5-MyD88-NLRP3 inflammasome. These findings highlight the urgent need for intervention strategies to mitigate the detrimental effects of air pollution on intestinal health and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Zihan Ran
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Jingcheng Yang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China; Greater Bay Area Institute of Precision Medicine, 115 Jiaoxi Road, Guangzhou 511458, China
| | - Liang Liu
- Clinical Research Unit, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shaobo Wu
- Department of Laboratory Medicine, Tinglin Hospital of Jinshan District, No. 80 Siping North Road, Shanghai 201505, China
| | - YanPeng An
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Wanwan Hou
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Tianyuan Cheng
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Youyi Zhang
- School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yiqing Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yechao Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Qianyue Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai 200011, China
| | - Jiaping Wan
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai 200011, China
| | - Xuemei Li
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Baoling Xing
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Yuchen Ye
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Penghao Xu
- School of Biological Sciences, Georgia Insitute of Technology, Atlanta, GA, USA
| | - Zhenghu Chen
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China.
| | - Jinzhuo Zhao
- School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China.
| | - Rui Li
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai 200011, China.
| |
Collapse
|
3
|
Ziki RA, Colnot S. Glutamine metabolism, a double agent combating or fuelling hepatocellular carcinoma. JHEP Rep 2024; 6:101077. [PMID: 38699532 PMCID: PMC11063524 DOI: 10.1016/j.jhepr.2024.101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 05/05/2024] Open
Abstract
The reprogramming of glutamine metabolism is a key event in cancer more generally and in hepatocellular carcinoma (HCC) in particular. Glutamine consumption supplies tumours with ATP and metabolites through anaplerosis of the tricarboxylic acid cycle, while glutamine production can be enhanced by the overexpression of glutamine synthetase. In HCC, increased glutamine production is driven by activating mutations in the CTNNB1 gene encoding β-catenin. Increased glutamine synthesis or utilisation impacts tumour epigenetics, oxidative stress, autophagy, immunity and associated pathways, such as the mTOR (mammalian target of rapamycin) pathway. In this review, we will discuss studies which emphasise the pro-tumoral or tumour-suppressive effect of glutamine overproduction. It is clear that more comprehensive studies are needed as a foundation from which to develop suitable therapies targeting glutamine metabolic pathways, depending on the predicted pro- or anti-tumour role of dysregulated glutamine metabolism in distinct genetic contexts.
Collapse
Affiliation(s)
- Razan Abou Ziki
- INSERM, Sorbonne Université, Centre de Recherche des Cordeliers (CRC), Paris, F-75006, France
- Équipe labellisée Ligue Nationale Contre le Cancer, France
| | - Sabine Colnot
- INSERM, Sorbonne Université, Centre de Recherche des Cordeliers (CRC), Paris, F-75006, France
- Équipe labellisée Ligue Nationale Contre le Cancer, France
| |
Collapse
|
4
|
Peehl DM, Badea CT, Chenevert TL, Daldrup-Link HE, Ding L, Dobrolecki LE, Houghton AM, Kinahan PE, Kurhanewicz J, Lewis MT, Li S, Luker GD, Ma CX, Manning HC, Mowery YM, O’Dwyer PJ, Pautler RG, Rosen MA, Roudi R, Ross BD, Shoghi KI, Sriram R, Talpaz M, Wahl RL, Zhou R. Animal Models and Their Role in Imaging-Assisted Co-Clinical Trials. Tomography 2023; 9:657-680. [PMID: 36961012 PMCID: PMC10037611 DOI: 10.3390/tomography9020053] [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: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
The availability of high-fidelity animal models for oncology research has grown enormously in recent years, enabling preclinical studies relevant to prevention, diagnosis, and treatment of cancer to be undertaken. This has led to increased opportunities to conduct co-clinical trials, which are studies on patients that are carried out parallel to or sequentially with animal models of cancer that mirror the biology of the patients' tumors. Patient-derived xenografts (PDX) and genetically engineered mouse models (GEMM) are considered to be the models that best represent human disease and have high translational value. Notably, one element of co-clinical trials that still needs significant optimization is quantitative imaging. The National Cancer Institute has organized a Co-Clinical Imaging Resource Program (CIRP) network to establish best practices for co-clinical imaging and to optimize translational quantitative imaging methodologies. This overview describes the ten co-clinical trials of investigators from eleven institutions who are currently supported by the CIRP initiative and are members of the Animal Models and Co-clinical Trials (AMCT) Working Group. Each team describes their corresponding clinical trial, type of cancer targeted, rationale for choice of animal models, therapy, and imaging modalities. The strengths and weaknesses of the co-clinical trial design and the challenges encountered are considered. The rich research resources generated by the members of the AMCT Working Group will benefit the broad research community and improve the quality and translational impact of imaging in co-clinical trials.
Collapse
Affiliation(s)
- Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Cristian T. Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Thomas L. Chenevert
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
| | - Heike E. Daldrup-Link
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA; (H.E.D.-L.); (R.R.)
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - Lacey E. Dobrolecki
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA;
| | | | - Paul E. Kinahan
- Department of Radiology, University of Washington, Seattle, WA 98105, USA;
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Michael T. Lewis
- Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - Gary D. Luker
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Cynthia X. Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - H. Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708, USA;
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC 27708, USA
| | - Peter J. O’Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
| | - Robia G. Pautler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Mark A. Rosen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA; (H.E.D.-L.); (R.R.)
| | - Brian D. Ross
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
- Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Kooresh I. Shoghi
- Mallinckrodt Institute of Radiology (MIR), Washington University School of Medicine, St. Louis, MO 63110, USA; (K.I.S.); (R.L.W.)
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Moshe Talpaz
- Division of Hematology/Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA;
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Richard L. Wahl
- Mallinckrodt Institute of Radiology (MIR), Washington University School of Medicine, St. Louis, MO 63110, USA; (K.I.S.); (R.L.W.)
| | - Rong Zhou
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Glutamine increases stability of TPH1 mRNA via p38 mitogen-activated kinase in mouse mastocytoma cells. Mol Biol Rep 2023; 50:267-277. [PMID: 36331742 PMCID: PMC9884262 DOI: 10.1007/s11033-022-07693-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/06/2022]
Abstract
Expression changes for tryptophan hydroxylase 1 (TPH1), the rate-limiting enzyme in serotonin synthesis, by environmental glutamine (GLN) were examined in mouse mastocytoma-derived P815-HTR cells. GLN-treated cells exhibited a robust increase in TPH1 mRNA after a 6 h exposure to GLN. 6-Diazo-5-oxo-L-norleucine (DON), a glutamine-utilizing glutaminase inhibitor, significantly inhibited the GLN-induction of TPH1 mRNA. Nuclear run-on assays and mRNA decay experiments demonstrated that the primary mechanism leading to increased TPH1 mRNA levels was not due to transcriptional changes, but rather due to increased TPH1 RNA stability induced by GLN. Treatment with GLN also led to activation of p38 MAP kinase, but not p42/44 MAPK. In addition, SB203580, a p38 MAP kinase specific inhibitor, completely abolished the GLN-mediated increase of TPH1 mRNA levels, suggesting the pathway stabilizing TPH1 mRNA might be mediated by the activated p38 MAP kinase pathway. Additionally, SB203580 significantly reduced the stability of TPH1 mRNA, and this reduction of the stability was not affected by GLN in the culture medium, implying a sequential signaling from GLN being mediated by p38 MAP kinase, resulting in alteration of TPH1 mRNA stability. TPH1 mRNA stability loss was also dependent on de novo protein synthesis as shown by treatment of cells with a transcriptional/translational blocker. We provide evidence that TPH1 mRNA levels are increased in response to increased exogenous GLN in mouse mastocytoma cells via a stabilization of TPH1 mRNA due to the activity of the p38 MAP kinase.
Collapse
|
6
|
Jiao C, Zou J, Chen Z, Zheng F, Xu Z, Lin YH, Wang Q. Dietary Glutamine Inclusion Regulates Immune and Antioxidant System, as Well as Programmed Cell Death in Fish to Protect against Flavobacterium columnare Infection. Antioxidants (Basel) 2021; 11:44. [PMID: 35052548 PMCID: PMC8773122 DOI: 10.3390/antiox11010044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/07/2023] Open
Abstract
The susceptibility of animals to pathogenic infection is significantly affected by nutritional status. The present study took yellow catfish (Pelteobagrus fulvidraco) as a model to test the hypothesis that the protective roles of glutamine during bacterial infection are largely related to its regulation on the immune and antioxidant system, apoptosis and autophagy. Dietary glutamine supplementation significantly improved fish growth performance and feed utilization. After a challenge with Flavobacterium columnare, glutamine supplementation promoted il-8 and il-1β expression via NF-κB signaling in the head kidney and spleen, but inhibited the over-inflammation in the gut and gills. Additionally, dietary glutamine inclusion also enhanced the systematic antioxidant capacity. Histological analysis showed the protective role of glutamine in gill structures. Further study indicated that glutamine alleviated apoptosis during bacterial infection, along with the reduced protein levels of caspase-3 and the reduced expression of apoptosis-related genes. Moreover, glutamine also showed an inhibitory role in autophagy which was due to the increased activation of the mTOR signaling pathway. Thus, our study for the first time illustrated the regulatory roles of glutamine in the fish immune and antioxidant system, and reported its inhibitory effects on fish apoptosis and autophagy during bacterial infection.
Collapse
Affiliation(s)
- Congrui Jiao
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Jiahong Zou
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Zhenwei Chen
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Feifei Zheng
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Zhen Xu
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| | - Yu-Hung Lin
- Department of Aquaculture, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 912, Taiwan
| | - Qingchao Wang
- College of Fisheries, Huazhong Agricultural University, 1 Shizishan Street, Wuhan 430070, China; (C.J.); (J.Z.); (Z.C.); (F.Z.); (Z.X.)
| |
Collapse
|
7
|
Choe JH, Mazambani S, Kim TH, Kim JW. Oxidative Stress and the Intersection of Oncogenic Signaling and Metabolism in Squamous Cell Carcinomas. Cells 2021; 10:606. [PMID: 33803326 PMCID: PMC8000417 DOI: 10.3390/cells10030606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Squamous cell carcinomas (SCCs) arise from both stratified squamous and non-squamous epithelium of diverse anatomical sites and collectively represent one of the most frequent solid tumors, accounting for more than one million cancer deaths annually. Despite this prevalence, SCC patients have not fully benefited from recent advances in molecularly targeted therapy or immunotherapy. Rather, decades old platinum-based or radiation regimens retaining limited specificity to the unique characteristics of SCC remain first-line treatment options. Historically, a lack of a consolidated perspective on genetic aberrations driving oncogenic transformation and other such factors essential for SCC pathogenesis and intrinsic confounding cellular heterogeneity in SCC have contributed to a critical dearth in effective and specific therapies. However, emerging evidence characterizing the distinct genomic, epigenetic, and metabolic landscapes of SCC may be elucidating unifying features in a seemingly heterogeneous disease. In this review, by describing distinct metabolic alterations and genetic drivers of SCC revealed by recent studies, we aim to establish a conceptual framework for a previously unappreciated network of oncogenic signaling, redox perturbation, and metabolic reprogramming that may reveal targetable vulnerabilities at their intersection.
Collapse
Affiliation(s)
- Joshua H. Choe
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Simbarashe Mazambani
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (S.M.); (T.H.K.)
| | - Tae Hoon Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (S.M.); (T.H.K.)
| | - Jung-whan Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (S.M.); (T.H.K.)
- Research and Development, VeraVerse Inc., 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
8
|
Host cell glutamine metabolism as a potential antiviral target. Clin Sci (Lond) 2021; 135:305-325. [PMID: 33480424 DOI: 10.1042/cs20201042] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022]
Abstract
A virus minimally contains a nucleic acid genome packaged by a protein coat. The genome and capsid together are known as the nucleocapsid, which has an envelope containing a lipid bilayer (mainly phospholipids) originating from host cell membranes. The viral envelope has transmembrane proteins that are usually glycoproteins. The proteins in the envelope bind to host cell receptors, promoting membrane fusion and viral entry into the cell. Virus-infected host cells exhibit marked increases in glutamine utilization and metabolism. Glutamine metabolism generates ATP and precursors for the synthesis of macromolecules to assemble progeny viruses. Some compounds derived from glutamine are used in the synthesis of purines and pyrimidines. These latter compounds are precursors for the synthesis of nucleotides. Inhibitors of glutamine transport and metabolism are potential candidate antiviral drugs. Glutamine is also an essential nutrient for the functions of leukocytes (lymphocyte, macrophage, and neutrophil), including those in virus-infected patients. The increased glutamine requirement for immune cell functions occurs concomitantly with the high glutamine utilization by host cells in virus-infected patients. The development of antiviral drugs that target glutamine metabolism must then be specifically directed at virus-infected host cells to avoid negative effects on immune functions. Therefore, the aim of this review was to describe the landscape of cellular glutamine metabolism to search for potential candidates to inhibit glutamine transport or glutamine metabolism.
Collapse
|
9
|
Amino Acids in Cell Signaling: Regulation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1332:17-33. [PMID: 34251636 DOI: 10.1007/978-3-030-74180-8_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amino acids are the main building blocks for life. Aside from their roles in composing proteins, functional amino acids and their metabolites play regulatory roles in key metabolic cascades, gene expressions, and cell-to-cell communication via a variety of cell signaling pathways. These metabolic networks are necessary for maintenance, growth, reproduction, and immunity in humans and animals. These amino acids include, but are not limited to, arginine, glutamine, glutamate, glycine, leucine, proline, and tryptophan. We will discuss these functional amino acids in cell signaling pathways in mammals with a particular emphasis on mTORC1, AMPK, and MAPK pathways for protein synthesis, nutrient sensing, and anti-inflammatory responses, as well as cell survival, growth, and development.
Collapse
|
10
|
Lin C, Salzillo TC, Bader DA, Wilkenfeld SR, Awad D, Pulliam TL, Dutta P, Pudakalakatti S, Titus M, McGuire SE, Bhattacharya PK, Frigo DE. Prostate Cancer Energetics and Biosynthesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:185-237. [PMID: 31900911 PMCID: PMC8096614 DOI: 10.1007/978-3-030-32656-2_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancers must alter their metabolism to satisfy the increased demand for energy and to produce building blocks that are required to create a rapidly growing tumor. Further, for cancer cells to thrive, they must also adapt to an often changing tumor microenvironment, which can present new metabolic challenges (ex. hypoxia) that are unfavorable for most other cells. As such, altered metabolism is now considered an emerging hallmark of cancer. Like many other malignancies, the metabolism of prostate cancer is considerably different compared to matched benign tissue. However, prostate cancers exhibit distinct metabolic characteristics that set them apart from many other tumor types. In this chapter, we will describe the known alterations in prostate cancer metabolism that occur during initial tumorigenesis and throughout disease progression. In addition, we will highlight upstream regulators that control these metabolic changes. Finally, we will discuss how this new knowledge is being leveraged to improve patient care through the development of novel biomarkers and metabolically targeted therapies.
Collapse
Affiliation(s)
- Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Travis C Salzillo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - David A Bader
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Dominik Awad
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Thomas L Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Prasanta Dutta
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Titus
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean E McGuire
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Molecular Medicine Program, The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
11
|
Schulte ML, Fu A, Zhao P, Li J, Geng L, Smith ST, Kondo J, Coffey RJ, Johnson MO, Rathmell JC, Sharick JT, Skala MC, Smith JA, Berlin J, Washington MK, Nickels ML, Manning HC. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat Med 2018; 24:194-202. [PMID: 29334372 PMCID: PMC5803339 DOI: 10.1038/nm.4464] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 11/29/2017] [Indexed: 12/11/2022]
Abstract
The unique metabolic demands of cancer cells underscore potentially fruitful opportunities for drug discovery in the era of precision medicine. However, therapeutic targeting of cancer metabolism has led to surprisingly few new drugs to date. The neutral amino acid glutamine serves as a key intermediate in numerous metabolic processes leveraged by cancer cells, including biosynthesis, cell signaling, and oxidative protection. Herein we report the preclinical development of V-9302, a competitive small molecule antagonist of transmembrane glutamine flux that selectively and potently targets the amino acid transporter ASCT2. Pharmacological blockade of ASCT2 with V-9302 resulted in attenuated cancer cell growth and proliferation, increased cell death, and increased oxidative stress, which collectively contributed to antitumor responses in vitro and in vivo. This is the first study, to our knowledge, to demonstrate the utility of a pharmacological inhibitor of glutamine transport in oncology, representing a new class of targeted therapy and laying a framework for paradigm-shifting therapies targeting cancer cell metabolism.
Collapse
Affiliation(s)
- Michael L. Schulte
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Allie Fu
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Ping Zhao
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jun Li
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Ling Geng
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Shannon T. Smith
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jumpei Kondo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, TN, 37212, United States
| | - Marc O. Johnson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Joe T. Sharick
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Jarrod A. Smith
- Vanderbilt Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Jordan Berlin
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - M. Kay Washington
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Michael L. Nickels
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - H. Charles Manning
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| |
Collapse
|
12
|
|
13
|
Human cytomegalovirus-encoded miR-US4-1 promotes cell apoptosis and benefits discharge of infectious virus particles by targeting QARS. J Biosci 2017; 41:183-92. [PMID: 27240979 DOI: 10.1007/s12038-016-9605-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human cytomegalovirus (HCMV) can cause congenital diseases and opportunistic infections in immunocompromised individuals. Its functional proteins and microRNAs (miRNAs) facilitate efficient viral propagation by altering host cell behaviour. Identification of functional target genes of miRNAs is an important step in studies on HCMV pathogenesis. In this study, Glutaminyl-tRNA Synthetase (QARS), which could regulate signal transduction pathways for cellular apoptosis, was identified as a direct target of hcmv-miR-US4-1. Apoptosis assay revealed that as silence of QARS by ectopic expression of hcmv-miR-US4-1 and specific small interference RNA of QARS can promote cell apoptosis in HCMV-infected HELF cells. Moreover, viral growth curve assays showed that hcmv-miR-US4-1 benefits the discharge of infectious virus particles. However, silence of hcmv-miR-US4-1 by its specific inhibitor overturned these effects. These results imply that hcmv-miR-US4-1 might have the same effects during HCMV nature infection. In general, hcmv-miR-US4-1 may involve in promoting cell apoptosis and benefiting discharge of infectious virus particles via down-regulation of QARS in HCMV-infected HELF cells.
Collapse
|
14
|
Ligthart-Melis GC, van de Poll MCG, Dejong CHC, Boelens PG, Deutz NEP, van Leeuwen PAM. The Route of Administration (Enteral or Parenteral) Affects the Conversion of Isotopically Labeled L-[2-15N]Glutamine Into Citrulline and Arginine in Humans. JPEN J Parenter Enteral Nutr 2017; 31:343-48; discussion 349-50. [PMID: 17712141 DOI: 10.1177/0148607107031005343] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Glutamine exhibits numerous beneficial effects in experimental and clinical studies. It has been suggested that these effects may be partly mediated by the conversion of glutamine into citrulline and arginine. The intestinal metabolism of glutamine appears to be crucial in this pathway. The present study was designed to establish the effect of the feeding route, enteral or parenteral, on the conversion of exogenously administered glutamine into citrulline and arginine at an organ level in humans, with a focus on gut metabolism. METHODS Sixteen patients undergoing upper gastrointestinal surgery received an IV or enteral (EN) infusion of L-[2-(15)N]glutamine. Blood was sampled from a radial artery and from the portal and right renal vein. Amino acid concentrations and enrichments were measured, and net fluxes of [(15)N]-labeled substrates across the portal drained viscera (PDV) and kidneys were calculated from arteriovenous differences and plasma flow. RESULTS Arterial [(15)N]glutamine enrichments were significantly lower during enteral tracer infusion (tracer-to-tracee ratio [labeled vs unlabeled substrate, TTR%] IV: 6.66 +/- 0.35 vs EN: 3.04 +/- 0.45; p < .01), reflecting first-pass intestinal metabolism of glutamine during absorption. Compared with IV administration, enteral administration of the glutamine tracer resulted in a significantly higher intestinal fractional extraction of [(15)N]glutamine (IV: 0.15 +/- 0.03 vs EN: 0.44 +/- 0.08 micromol/kg/h; p < .01). Furthermore, enteral administration of the glutamine tracer resulted in higher arterial enrichments of [(15)N]citrulline (TTR% IV: 5.52 +/- 0.44 vs EN: 8.81 +/- 1.1; p = .02), and both routes of administration generated a significant enrichment of [(15)N]arginine (TTR% IV: 1.43 +/- 0.12 vs EN: 1.68 +/- 0.18). This was accompanied by intestinal release of [(15)N]citrulline across the PDV, which was higher with enteral glutamine (IV: 0.38 +/- 0.07 vs EN: 0.72 +/- 0.11 micromol/kg/h; p = .02), and subsequent [(15)N]arginine release in both groups. CONCLUSIONS In humans, the gut preferably takes up enterally administered glutamine compared with intravenously provided glutamine. The route of administration, enteral or IV, affects the quantitative conversion of glutamine into citrulline and subsequent renal arginine synthesis in humans.
Collapse
|
15
|
Li DD, Zhong BW, Zhang HX, Zhou HY, Luo J, Liu Y, Xu GC, Luan CS, Fang J. Inhibition of the oxidative stress-induced miR-23a protects the human retinal pigment epithelium (RPE) cells from apoptosis through the upregulation of glutaminase and glutamine uptake. Mol Biol Rep 2016; 43:1079-87. [DOI: 10.1007/s11033-016-4041-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 07/08/2016] [Indexed: 11/28/2022]
|
16
|
The glutamine-alpha-ketoglutarate (AKG) metabolism and its nutritional implications. Amino Acids 2016; 48:2067-80. [DOI: 10.1007/s00726-016-2254-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/04/2016] [Indexed: 01/08/2023]
|
17
|
Moore SR, Guedes MM, Costa TB, Vallance J, Maier EA, Betz KJ, Aihara E, Mahe MM, Lima AAM, Oriá RB, Shroyer NF. Glutamine and alanyl-glutamine promote crypt expansion and mTOR signaling in murine enteroids. Am J Physiol Gastrointest Liver Physiol 2015; 308:G831-9. [PMID: 25792564 PMCID: PMC4437023 DOI: 10.1152/ajpgi.00422.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/13/2015] [Indexed: 01/31/2023]
Abstract
L-glutamine (Gln) is a key metabolic fuel for intestinal epithelial cell proliferation and survival and may be conditionally essential for gut homeostasis during catabolic states. We show that L-alanyl-L-glutamine (Ala-Gln), a stable Gln dipeptide, protects mice against jejunal crypt depletion in the setting of dietary protein and fat deficiency. Separately, we show that murine crypt cultures (enteroids) derived from the jejunum require Gln or Ala-Gln for maximal expansion. Once expanded, enteroids deprived of Gln display a gradual atrophy of cryptlike domains, with decreased epithelial proliferation, but stable proportions of Paneth and goblet cell differentiation, at 24 h. Replenishment of enteroid medium with Gln selectively activates mammalian target of rapamycin (mTOR) signaling pathways, rescues proliferation, and promotes crypt regeneration. Gln deprivation beyond 48 h leads to destabilization of enteroids but persistence of EGFP-Lgr5-positive intestinal stem cells with the capacity to regenerate enteroids upon Gln rescue. Collectively, these findings indicate that Gln deprivation induces a reversible quiescence of intestinal stem cells and provides new insights into nutritional regulation of intestinal epithelial homeostasis.
Collapse
Affiliation(s)
- Sean R. Moore
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio;
| | - Marjorie M. Guedes
- 2Department of Physiology and Pharmacology, Clinical Research Unit and Institute of Biomedicine/Center for Global Health, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil;
| | - Tie B. Costa
- 3Department of Morphology, Clinical Research Unit and Institute of Biomedicine/Center for Global Health, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil;
| | - Jefferson Vallance
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio;
| | - Elizabeth A. Maier
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio;
| | - Kristina J. Betz
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio;
| | - Eitaro Aihara
- 4Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| | - Maxime M. Mahe
- 5Division of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Aldo A. M. Lima
- 2Department of Physiology and Pharmacology, Clinical Research Unit and Institute of Biomedicine/Center for Global Health, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil;
| | - Reinaldo B. Oriá
- 2Department of Physiology and Pharmacology, Clinical Research Unit and Institute of Biomedicine/Center for Global Health, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil;
| | - Noah F. Shroyer
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio;
| |
Collapse
|
18
|
Wang W, Wu Z, Lin G, Hu S, Wang B, Dai Z, Wu G. Glycine stimulates protein synthesis and inhibits oxidative stress in pig small intestinal epithelial cells. J Nutr 2014; 144:1540-8. [PMID: 25122646 DOI: 10.3945/jn.114.194001] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glycine has recently been classified as a nutritionally essential amino acid for maximal growth in young pigs. Currently, little is known about the metabolism or function of glycine in the neonatal intestine. This work was conducted to test the hypothesis that glycine has a protective effect against oxidative stress in intestinal epithelial cells. Jejunal enterocytes isolated from newborn pigs were cultured in the presence of 0.0-2 mmol/L glycine for measurements of glycine metabolism, cell proliferation, protein turnover, apoptosis, and antioxidative response. Compared with 0.0-0.5 mmol/L glycine, 1.0 mmol/L glycine enhanced (P < 0.05) cell growth (by 8-24% on day 2 and by 34-224% on day 4, respectively) and protein synthesis (by 36-419%) while reducing (P < 0.05) protein degradation (by 7-28%). This effect of glycine was associated with activation of the mammalian target of rapamycin signaling pathway in enterocytes. By using a model of oxidative stress induced by 30 μmol/L 4-hydroxynonenal (4-HNE), which was assessed by flow cytometry analysis, 1.0 mmol/L glycine inhibited (P < 0.05) activation of caspase 3 by 25% and attenuated (P < 0.05) 4-HNE-induced apoptosis by 38% in intestinal porcine epithelial cell line 1 cells through promotion of reduced glutathione synthesis and expression of glycine transporter 1 while reducing the activation of extracellular signal-regulated kinases, c-Jun amino-terminal kinases, and p38 protein in the mitogen-activated protein kinase signaling pathway. These novel findings provide a biochemical mechanism for the use of dietary glycine to improve intestinal health in neonates under conditions of oxidative stress and glycine deficiency.
Collapse
Affiliation(s)
- Weiwei Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China; and Department of Animal Science, Texas A&M University, College Station, TX
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China; and
| | - Gang Lin
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China; and
| | - Shengdi Hu
- Department of Animal Science, Texas A&M University, College Station, TX
| | - Bin Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China; and
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China; and
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China; and Department of Animal Science, Texas A&M University, College Station, TX
| |
Collapse
|
19
|
Maretta M, Tóth Š, Jonecová Z, Veselá J. Impact of alanyl-glutamine dipeptide on proliferative and inflammatory changes in jejunal mucosa after acute mesenteric ischemia. J Pediatr Surg 2014; 49:1385-9. [PMID: 25148743 DOI: 10.1016/j.jpedsurg.2014.01.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/09/2013] [Accepted: 01/03/2014] [Indexed: 02/02/2023]
Abstract
PURPOSE The aim of our study was to determinate the impact of dipeptide (alanyl-glutamine) administration on inflammatory and proliferative changes in jejunal mucosa after acute mesenteric ischemia. METHODS Male Wistar rats (n=30) were divided into three groups: ischemia/reperfusion (IR) group which undergoes 60min of mesenteric ischemia and 1 or 24h of reperfusion (IR1, IR24, n=12). Groups with dipeptide administration (D+IR1, D+IR24, Dipeptiven con inf., i.v., 0.75 g/kg) prior to IR injury were followed by 1 and 24h of reperfusion. At the end of reperfusion period jejunal bioptic samples were obtained for histological (H&E), histochemical (Alcian blue) and immunohistochemical (anti-PCNA, anti-MPO) evaluations. RESULTS Our results pointed out a significant (p<0.001) increase of histopathological injury score in IR1 group compared to D+IR1 group. Immunohistochemical evaluation showed that MPO-positivity was significantly increased in IR groups after 1 (p<0.001) as well as 24h of reperfusion (p<0.01) compared to dipeptide pretreated groups. Proliferative/reparatory rate was assessed using anti-PCNA antibody and showed a significant increase (p<0.01) in PCNA cell positivity in lamina propria in dipeptide treated group compared to IR group. CONCLUSION In conclusion we may suggest that administration of alanyl-glutamine dipeptide prior to IR injury may help to protect small intestine and its mucous membrane integrity against insult such as intestinal ischemic/reperfusion injury presents.
Collapse
Affiliation(s)
- Milan Maretta
- Department of Histology and Embryology, Faculty of Medicine, Pavol Jozef Šafárik University, Šrobárova 2, Košice, Slovak Republic.
| | - Štefan Tóth
- Department of Histology and Embryology, Faculty of Medicine, Pavol Jozef Šafárik University, Šrobárova 2, Košice, Slovak Republic
| | - Zuzana Jonecová
- Department of Histology and Embryology, Faculty of Medicine, Pavol Jozef Šafárik University, Šrobárova 2, Košice, Slovak Republic
| | - Jarmila Veselá
- Department of Histology and Embryology, Faculty of Medicine, Pavol Jozef Šafárik University, Šrobárova 2, Košice, Slovak Republic
| |
Collapse
|
20
|
Zhu Y, Lin G, Dai Z, Zhou T, Li T, Yuan T, Wu Z, Wu G, Wang J. L-Glutamine deprivation induces autophagy and alters the mTOR and MAPK signaling pathways in porcine intestinal epithelial cells. Amino Acids 2014; 47:2185-97. [PMID: 24997162 DOI: 10.1007/s00726-014-1785-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022]
Abstract
L-Glutamine (Gln) is an essential amino acid for intestinal growth and integrity. However, the underlying molecular mechanisms are not fully known. In the present study, porcine intestinal epithelial cells (IPEC-1) were used to test the hypothesis that autophagy is induced by Gln deprivation and inhibited by Gln supplementation. After a 2-day period of growth in normal medium, IPEC-1 cells were transferred to a Gln-free custom-made DMEM. Cell numbers, the distribution of autophagosomes, the abundance of the protein for an autophagy marker LC3B, as well as abundances of the mTOR and MAPK proteins during an 8-h period were determined. Furthermore, the rescue effect of 5 mM Gln was evaluated. Our results showed that Gln deprivation reduced the cell number, while enhancing the accumulation of autophagosomes and the expression of LC3B-II in IPEC-1 cells within 8 h. The concentrations of Glu, Asp, Cit, Arg, Leu, Ile, Val, Ala, β-Ala, Orn, Phe, Met and Ser in the culture medium were altered by Gln deprivation. Further analysis revealed that Gln deficiency inactivated, but Gln supplementation activated, the mTOR and MAPK/ERK signaling pathways. Collectively, our findings support the notion that Gln deficiency induces autophagy and disturbs amino acid metabolism in intestinal epithelial cells, as well as attenuated their mTOR and MAPK/ERK signaling pathways to inhibit protein synthesis and cell proliferation.
Collapse
Affiliation(s)
- Yuhua Zhu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Gang Lin
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Tianjiao Zhou
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Tiantian Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Taolin Yuan
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.,Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
21
|
Dietary requirements of "nutritionally non-essential amino acids" by animals and humans. Amino Acids 2012; 44:1107-13. [PMID: 23247926 DOI: 10.1007/s00726-012-1444-2] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 12/02/2012] [Indexed: 01/08/2023]
Abstract
Amino acids are necessary for the survival, growth, development, reproduction and health of all organisms. They were traditionally classified as nutritionally essential or non-essential for mammals, birds and fish based on nitrogen balance or growth. It was assumed that all "non-essential amino acids (NEAA)" were synthesized sufficiently in the body to meet the needs for maximal growth and health. However, there has been no compelling experimental evidence to support this assumption over the past century. NEAA (e.g., glutamine, glutamate, proline, glycine and arginine) play important roles in regulating gene expression, cell signaling, antioxidative responses, neurotransmission, and immunity. Additionally, glutamate, glutamine and aspartate are major metabolic fuels for the small intestine to maintain its digestive function and protect its mucosal integrity. Therefore, based on new research findings, NEAA should be taken into consideration in revising the classical "ideal protein" concept and formulating balanced diets to improve protein accretion, food efficiency, and health in animals and humans.
Collapse
|
22
|
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
|
23
|
Lesueur C, Bôle-Feysot C, Bekri S, Husson A, Lavoinne A, Brasse-Lagnel C. Glutamine induces nuclear degradation of the NF-κB p65 subunit in Caco-2/TC7 cells. Biochimie 2012; 94:806-15. [DOI: 10.1016/j.biochi.2011.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 11/22/2011] [Indexed: 12/22/2022]
|
24
|
Mohammad S, Ramos LS, Buck J, Levin LR, Rubino F, McGraw TE. Gastric inhibitory peptide controls adipose insulin sensitivity via activation of cAMP-response element-binding protein and p110β isoform of phosphatidylinositol 3-kinase. J Biol Chem 2011; 286:43062-70. [PMID: 22027830 DOI: 10.1074/jbc.m111.289009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gastric inhibitory peptide (GIP) is an incretin hormone secreted in response to food intake. The best known function of GIP is to enhance glucose-dependent insulin secretion from pancreatic β-cells. Extra-pancreatic effects of GIP primarily occur in adipose tissues. Here, we demonstrate that GIP increases insulin-dependent translocation of the Glut4 glucose transporter to the plasma membrane and exclusion of FoxO1 transcription factor from the nucleus in adipocytes, establishing that GIP has a general effect on insulin action in adipocytes. Stimulation of adipocytes with GIP alone has no effect on these processes. Using pharmacologic and molecular genetic approaches, we show that the effect of GIP on adipocyte insulin sensitivity requires activation of both the cAMP/protein kinase A/CREB signaling module and p110β phosphoinositol-3' kinase, establishing a novel signal transduction pathway modulating insulin action in adipocytes. This insulin-sensitizing effect is specific for GIP because isoproterenol, which elevates adipocyte cAMP and activates PKA/CREB signaling, does not affect adipocyte insulin sensitivity. The insulin-sensitizing activity points to a more central role for GIP in intestinal regulation of peripheral tissue metabolism, an emerging feature of inter-organ communication in the control of metabolism.
Collapse
Affiliation(s)
- Sameer Mohammad
- Department of Biochemistry, Weill Medical College of Cornell University, New York, New York 10065, USA
| | | | | | | | | | | |
Collapse
|
25
|
Ueno PM, Oriá RB, Maier EA, Guedes M, de Azevedo OG, Wu D, Willson T, Hogan SP, Lima AAM, Guerrant RL, Polk DB, Denson LA, Moore SR. Alanyl-glutamine promotes intestinal epithelial cell homeostasis in vitro and in a murine model of weanling undernutrition. Am J Physiol Gastrointest Liver Physiol 2011; 301:G612-22. [PMID: 21799183 PMCID: PMC3191556 DOI: 10.1152/ajpgi.00531.2010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alanyl-glutamine (Ala-Gln) has recently been shown to enhance catch-up growth and gut integrity in undernourished children from Northeast Brazil. We hypothesized that the intestinal epithelial effects of Ala-Gln in malnourished weanling mice and mouse small intestinal epithelial (MSIE) cells would include modulation of barrier function, proliferation, and apoptosis. Dams of 10-day-old suckling C57BL/6 pups were randomized to a standard diet or an isocaloric Northeast Brazil "regional basic diet," moderately deficient in protein, fat, and minerals. Upon weaning to their dam's diet on day of life 21, pups were randomized to Ala-Gln solution or water. At 6 wk of age, mice were killed, and jejunal tissue was collected for morphology, immunohistochemistry, and Ussing chamber analysis of transmucosal resistance and permeability. Proliferation of MSIE cells in the presence or absence of Ala-Gln was measured by MTS and bromodeoxyuridine assays. MSIE apoptosis was assessed by annexin and 7-amino-actinomycin D staining. Pups of regional basic diet-fed dams exhibited failure to thrive. Jejunal specimens from undernourished weanlings showed decreased villous height and crypt depth, decreased transmucosal resistance, increased permeability to FITC-dextran, increased claudin-3 expression, and decreased epithelial proliferation and increased epithelial apoptosis (as measured by bromodeoxyuridine and cleaved caspase-3 staining, respectively). Undernourished weanlings supplemented with Ala-Gln showed improvements in weight velocity, villous height, crypt depth, transmucosal resistance, and epithelial proliferation/apoptosis compared with unsupplemented controls. Similarly, Ala-Gln increased proliferation and reduced apoptosis in MSIE cells. In summary, Ala-Gln promotes intestinal epithelial homeostasis in a mouse model of malnutrition-associated enteropathy, mimicking key features of the human disease.
Collapse
Affiliation(s)
- Priscilla M. Ueno
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | | | - Elizabeth A. Maier
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Marjorie Guedes
- 3Institute of Biomedicine, Federal University of Ceará, Ceará, Brazil;
| | | | - David Wu
- 4Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Tara Willson
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Simon P. Hogan
- 4Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Aldo A. M. Lima
- 3Institute of Biomedicine, Federal University of Ceará, Ceará, Brazil;
| | - Richard L. Guerrant
- 5Center for Global Health, University of Virginia, Charlottesville, Virginia; and
| | - D. Brent Polk
- 6Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Lee A. Denson
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Sean R. Moore
- 1Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| |
Collapse
|
26
|
Nakamura A, Hara K, Yamamoto K, Yasuda H, Moriyama H, Hirai M, Nagata M, Yokono K. Role of the mTOR complex 1 pathway in the in vivo maintenance of the intestinal mucosa by oral intake of amino acids. Geriatr Gerontol Int 2011; 12:131-9. [PMID: 21794051 DOI: 10.1111/j.1447-0594.2011.00729.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIM Oral intake of nutrients is often compromised in elderly, multimorbid patients, but parenteral nutrition causes intestinal atrophy and impairs intestinal function. To uncover the molecular mechanisms by which amino acids are involved in intestinal atrophy and recovery, we studied whether the rapamycin-sensitive mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) pathway is involved in this process. METHODS C57BL/6N mice were fed a glucose solution alone, glucose solution with amino acids or normal chow diet for various lengths of time. Intestinal sections were prepared from these mice and the villus height and villus density were quantified. As a readout for the mTORC1 pathway, the phosphorylation of the ribosomal S6 protein (S6) was analyzed by immunostaining and immunoblotting. To confirm the role of the mTORC1 pathway, the inhibitory effect of a specific mTOR inhibitor, rapamycin, was examined. RESULTS Inducing fasting in mice fed only glucose caused time-dependent intestinal mucosal atrophy, whereas supplementation with amino acids protected the intestinal mucosa from atrophy. Phosphorylation of S6 decreased in the intestinal mucosa of mice fed only glucose, whereas supplementation with amino acids increased S6 phosphorylation. Importantly, intraperitoneal injection of rapamycin attenuated the protective effect of amino acids on the intestinal mucosa in a pattern consistent with the decrease of S6 phosphorylation. CONCLUSIONS These results indicate that the mTORC1 pathway plays a crucial role in the in vivo maintenance of the intestinal mucosa by the oral intake of amino acids.
Collapse
Affiliation(s)
- Akira Nakamura
- Department of Internal and Geriatric Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ban K, Kozar RA. Glutamine protects against apoptosis via downregulation of Sp3 in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2010; 299:G1344-53. [PMID: 20884886 PMCID: PMC3006244 DOI: 10.1152/ajpgi.00334.2010] [Citation(s) in RCA: 22] [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
Glutamine plays a key role in intestinal growth and maintenance of gut function, and as we have shown protects the postischemic gut (Kozar RA, Scultz SG, Bick RJ, Poindexter BJ, Desoigne R, Weisbrodt NW, Haber MM, Moore FA. Shock 21: 433-437, 2004). However, the precise mechanisms of the gut protective effects of glutamine have not been well elucidated. In the present study, RNA microarray was performed to obtain differentially expressed genes in intestinal epithelial IEC-6 cells following either 2 mM or 10 mM glutamine. The result demonstrated that specificity protein 3 (Sp3) mRNA expression was downregulated 3.1-fold. PCR and Western blot confirmed that Sp3 expression was decreased by glutamine in a time- and dose-dependent fashion. To investigate the role of Sp3, Sp3 gene siRNA silencing was performed and apoptosis was assessed. Silencing of Sp3 demonstrated a significant increase in Bcl-2 and decrease in Bax protein expression, as well as a decrease in caspase-3, -8, and -9 protein expression and activity. The protein expression of apoptosis-related proteins after hypoxia/reoxygenation was similar to that of normoxia and correlated with a decrease in DNA fragmentation. Importantly, the addition of glutamine to Sp3-silenced cells did not further lessen apoptosis, suggesting that Sp3 plays a major role in the inhibitory effect of glutamine on apoptosis. This novel finding may explain in part the gut-protective effects of glutamine.
Collapse
Affiliation(s)
- Kechen Ban
- Department of Surgery, University of Texas Medical School, Houston, 77030, USA.
| | | |
Collapse
|
28
|
Coëffier M, Marion-Letellier R, Déchelotte P. Potential for amino acids supplementation during inflammatory bowel diseases. Inflamm Bowel Dis 2010; 16:518-24. [PMID: 19572337 DOI: 10.1002/ibd.21017] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathophysiology of inflammatory bowel diseases (IBDs) is multifactorial and involves interactions of gut luminal content with mucosal barrier and especially immune cells. Malnutrition is a frequent issue during IBD flares, especially in Crohn's disease (CD) patients, and nutritional support is frequently used to treat malnutrition but also in an attempt to modulate intestinal inflammation. The use of oral or enteral nutrition intervention in IBDs may be effective, alone or in combination with drugs, to achieve and maintain remission. However, standard diets are less effective than new-generation biotherapies and could be improved by supplementation with specific immunomodulatory amino acids. Experimental studies evaluating glutamine, the preferential substrate for enterocytes, are promising. Some clinical studies with oral glutamine in CD are until now disappointing, but new formulations and targeting could enhance glutamine efficacy at the site of mucosal lesions. The role of arginine, involved in nitric oxide and polyamines synthesis, still remains debated. However, the effects of these amino acids in IBD have been poorly documented in humans. Other candidates like glycine, cysteine, histidine, or taurine should also be evaluated in the future.
Collapse
Affiliation(s)
- Moïse Coëffier
- Appareil Digestif Environnement Nutrition (ADEN EA4311), Institute for Biomedical Research, European Institute for Peptide Research (IFRMP 23), Rouen University and Rouen University Hospital, Rouen, France.
| | | | | |
Collapse
|
29
|
Brasse-Lagnel CG, Lavoinne AM, Husson AS. Amino acid regulation of mammalian gene expression in the intestine. Biochimie 2010; 92:729-35. [PMID: 20188788 DOI: 10.1016/j.biochi.2010.02.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 02/16/2010] [Indexed: 12/16/2022]
Abstract
Some amino acids exert a wide range of regulatory effects on gene expression via the activation of different signalling pathways and transcription factors, and a number of cis elements were shown to respond to changes in amino acid concentration. Particular attention has been paid to the effects of glutamine and arginine, which modulate a number of cell functions through the activation of various pathways in different tissues. In the intestine, appropriate concentrations of both arginine and/or glutamine contribute to facilitate cell proliferation, to limit the inflammatory response and apoptosis, and to modulate intermediary metabolism through specific transcription factors. Particularly, besides its role as a major fuel for enterocytes, the regulatory effects of glutamine have been extensively studied and the molecular mechanisms involved appear diversified and complex. Indeed, in addition to a major role of NF-kappaB in its anti-inflammatory action and a stimulatory role of AP-1 in its growth-promoting action and cell survival, the involvement of some other transcription factors, such as PPAR-gamma or HSF-1, was shown to maintain intestinal cell integrity. The signalling pathways leading to the activation of transcription factors imply several kinases, particularly MAP kinases in the effect of glutamine and p70 S6 kinase for those of arginine, but in most cases the precise pathways from the entrance of the aminoacid into the cell to the activation of gene transcription has remained elusive.
Collapse
Affiliation(s)
- Carole G Brasse-Lagnel
- Appareil Digestif, Environnement et Nutrition (ADEN EA 4311), IFR n degrees 23, Université de Rouen, 22 boulevard Gambetta, Rouen cedex, France
| | | | | |
Collapse
|
30
|
Taylor J, Cooper C, Mommsen T. Implications of GI function for gas exchange, acid–base balance and nitrogen metabolism. FISH PHYSIOLOGY 2010. [DOI: 10.1016/s1546-5098(10)03006-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
|
32
|
Domeneghini C, Di Giancamillo A, Bosi G, Arrighi S. Can nutraceuticals affect the structure of intestinal mucosa? Qualitative and quantitative microanatomy in L-glutamine diet-supplemented weaning piglets. Vet Res Commun 2009; 30:331-42. [PMID: 16437309 DOI: 10.1007/s11259-006-3236-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2004] [Indexed: 01/16/2023]
Abstract
Weaning piglets were fed an L-glutamine-supplemented diet with the aim of monitoring the effects on gut mucosal turnover and barrier function, to elucidate the potential preventive or therapeutic roles of glutamine as a nutraceutical or 'functional food'. Sixteen female weaning piglets were divided into two groups, which were fed a control diet (Ctr group: n = 8) or a Ctr + 0.5% L-glutamine diet (G group: n = 8) for 28 days. In the ileum of group G piglets the villus height (V) and crypt depth (C) were increased, and the V:C ratio was decreased (p < 0.01). The PCNA and TUNEL immunoreactivities were also tested. The number of mitotic mucosal cells (M) was increased, and that of mucosal cells with apoptotic nuclei (A) decreased in such a way that the A:M index diminished (p < 0.01). The A:M index also decreased at the level of some components of the gut-associated lymphatic tissue (GALT), thus indicating a positive effect on the gut barrier function. This trial showed that L: -glutamine supplementation influenced some morphofunctional characteristics of piglet ileal mucosa. These data corroborate the nutraceutical role of glutamine as a trophic agent for mucosal repair, improvement of barrier function and gut adaptation in the swine per se and as an animal model.
Collapse
Affiliation(s)
- C Domeneghini
- Department of Veterinary Sciences and Technologies for Food Safety, University of Milan, Via Trentacoste 2, Milan, I-20134, Italy.
| | | | | | | |
Collapse
|
33
|
Abstract
This review deals with the complications and treatment of short bowel syndrome (SBS), addressing the psychosocial, medical, and surgical complications in children receiving long-term parenteral nutrition (PN) support, as well as factors that affect the intestinal adaptation process. Whenever possible, as much of the colon as possible is retained in continuity because the colon is an avid absorber of sodium. It is also important for clinicians to be aware of the important absorption mechanisms in the different regions of the bowel. For example, resection of the terminal ileum removes vitamin B12 transporters and active sodium-coupled bile salt transporters. Treatment of patients missing the terminal ileum may require monthly vitamin B12 injections and oral bile salt binders, such as cholestyramine, when the colon is present to reduce the volume of diarrhea. Patients who do not have ileocecal valves (ICVs) are prone to small bowel bacterial overgrowth that requires treatment to facilitate the intestinal adaptation process. We discuss how the PN is decreased as enteral feedings are advanced as well as clinical monitoring and routine laboratory tests. Although much has been learned over the past 20 years about PN, major questions remain, including determining the optimal form of intravenous lipid (omega-3 preparations versus omega-6 lipids versus a combination of both) to prevent liver disease.
Collapse
Affiliation(s)
- Fernando Navarro
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| | - Wallace A. Gleason
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| | - J. Marc Rhoads
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| | - Ruben E. Quiros-Tejeira
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| |
Collapse
|
34
|
Abstract
Short bowel syndrome (SBS) is a relatively common, often lethal, and highly costly medical problem in North America. Necrotizing enterocolitis (NEC) is the leading cause of SBS in the United States. An important fact to remember is that the length of the small bowel in a 28-week preterm infant is about 150 cm and in a term infant is about 250 cm. Twenty percent of this length is generally sufficient to allow dependence on parenteral nutrition (PN) via intestinal adaptation. This process is driven by significant increases in circulating trophic hormones, such as cholecystokinin, epidermal and keratinocyte growth factors, growth hormone, insulin-like growth factor-1, and glucagon-like peptide-2. These hormones produce hypertrophy and hyperplasia of the villi, along with increases in specific brush border membrane absorption mechanisms, such as glucose-sodium cotransport (via SGLT-1) and peptide transport (via Pep-T1). Currently, the best clinical markers of intestinal adaptation are the calculated percentage of enteral versus parenteral calories in a growing infant who has SBS and the serum concentrations of citrulline, an amino acid synthesized by mature enterocytes that has been used as a measure of functional intestinal mass.
Collapse
Affiliation(s)
- Fernando Navarro
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| | - Wallace A. Gleason
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| | - J. Marc Rhoads
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| | - Ruben E. Quiros-Tejeira
- The University of Texas Medical School at Houston and Department of Pediatrics, Children's Memorial Hermann Hospital, Houston, Tex
| |
Collapse
|
35
|
Marc Rhoads J, Wu G. Glutamine, arginine, and leucine signaling in the intestine. Amino Acids 2009; 37:111-122. [PMID: 19130170 DOI: 10.1007/s00726-008-0225-4] [Citation(s) in RCA: 234] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 12/09/2008] [Indexed: 12/14/2022]
Abstract
Glutamine and leucine are abundant constituents of plant and animal proteins, whereas the content of arginine in foods and physiological fluids varies greatly. Besides their role in protein synthesis, these three amino acids individually activate signaling pathway to promote protein synthesis and possibly inhibit autophagy-mediated protein degradation in intestinal epithelial cells. In addition, glutamine and arginine stimulate the mitogen-activated protein kinase and mammalian target of rapamycin (mTOR)/p70 (s6) kinase pathways, respectively, to enhance mucosal cell migration and restitution. Moreover, through the nitric oxide-dependent cGMP signaling cascade, arginine regulates multiple physiological events in the intestine that are beneficial for cell homeostasis and survival. Available evidence from both in vitro and in vivo animal studies shows that glutamine and arginine promote cell proliferation and exert differential cytoprotective effects in response to nutrient deprivation, oxidative injury, stress, and immunological challenge. Additionally, when nitric oxide is available, leucine increases the migration of intestinal cells. Therefore, through cellular signaling mechanisms, arginine, glutamine, and leucine play crucial roles in intestinal growth, integrity, and function.
Collapse
Affiliation(s)
- J Marc Rhoads
- Department of Pediatrics, University of Texas Medical School at Houston, Houston, TX 77030, USA.
| | | |
Collapse
|
36
|
Brasse-Lagnel C, Lavoinne A, Husson A. Control of mammalian gene expression by amino acids, especially glutamine. FEBS J 2009; 276:1826-44. [PMID: 19250320 DOI: 10.1111/j.1742-4658.2009.06920.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Molecular data rapidly accumulating on the regulation of gene expression by amino acids in mammalian cells highlight the large variety of mechanisms that are involved. Transcription factors, such as the basic-leucine zipper factors, activating transcription factors and CCAAT/enhancer-binding protein, as well as specific regulatory sequences, such as amino acid response element and nutrient-sensing response element, have been shown to mediate the inhibitory effect of some amino acids. Moreover, amino acids exert a wide range of effects via the activation of different signalling pathways and various transcription factors, and a number of cis elements distinct from amino acid response element/nutrient-sensing response element sequences were shown to respond to changes in amino acid concentration. Particular attention has been paid to the effects of glutamine, the most abundant amino acid, which at appropriate concentrations enhances a great number of cell functions via the activation of various transcription factors. The glutamine-responsive genes and the transcription factors involved correspond tightly to the specific effects of the amino acid in the inflammatory response, cell proliferation, differentiation and survival, and metabolic functions. Indeed, in addition to the major role played by nuclear factor-kappaB in the anti-inflammatory action of glutamine, the stimulatory role of activating protein-1 and the inhibitory role of C/EBP homology binding protein in growth-promotion, and the role of c-myc in cell survival, many other transcription factors are also involved in the action of glutamine to regulate apoptosis and intermediary metabolism in different cell types and tissues. The signalling pathways leading to the activation of transcription factors suggest that several kinases are involved, particularly mitogen-activated protein kinases. In most cases, however, the precise pathways from the entrance of the amino acid into the cell to the activation of gene transcription remain elusive.
Collapse
Affiliation(s)
- Carole Brasse-Lagnel
- Appareil Digestif, Environnement et Nutrition, EA 4311, Université de Rouen, France
| | | | | |
Collapse
|
37
|
Guerrant RL, Oriá RB, Moore SR, Oriá MOB, Lima AAM. Malnutrition as an enteric infectious disease with long-term effects on child development. Nutr Rev 2009; 66:487-505. [PMID: 18752473 DOI: 10.1111/j.1753-4887.2008.00082.x] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Malnutrition is a major contributor to mortality and is increasingly recognized as a cause of potentially lifelong functional disability. Yet, a rate-limiting step in achieving normal nutrition may be impaired absorptive function due to multiple repeated enteric infections. This is especially problematic in children whose diets are marginal. In malnourished individuals, the infections are even more devastating. This review documents the evidence that intestinal infections lead to malnutrition and that malnutrition worsens intestinal infections. The clinical data presented here derive largely from long-term cohort studies that are supported by controlled animal studies. Also reviewed are the mechanisms by which enteric infections lead to undernutrition and by which malnutrition worsens enteric infections, with implications for potential novel interventions. Further intervention studies are needed to document the relevance of these mechanisms and, most importantly, to interrupt the vicious diarrhea-malnutrition cycle so children may develop their full potential.
Collapse
Affiliation(s)
- Richard L Guerrant
- Department of Medicine, University of Virginia, Charlottesville 22901, USA.
| | | | | | | | | |
Collapse
|
38
|
|
39
|
Rhoads JM, Liu Y, Niu X, Surendran S, Wu G. Arginine stimulates cdx2-transformed intestinal epithelial cell migration via a mechanism requiring both nitric oxide and phosphorylation of p70 S6 kinase. J Nutr 2008; 138:1652-1657. [PMID: 18716165 DOI: 10.1093/jn/138.9.1652] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In intestinal cells, arginine (Arg) is 1 of the 2 most potent amino acid activators of p70(s6k), a key regulator of 5'- terminal oligopyrimidine mRNA translation, a necessary condition for increased cell migration. To investigate the mechanism of response to Arg, we used the rat crypt cell line cdx2-transformed IEC-6 cells (cdx2-IEC) and measured cell migration, immunocytochemical analysis of p70(s6k) activation in response to Arg, and production of nitric oxide (NO). When treated with Arg, cdx2-IEC increased in phosphorylation on Thr-389 of p70(s6k) (pp70(s6k)) compared with control (P < 0.01). Phospho-Thr-421/Ser-424-p70(s6k) was located in the nucleus shortly after Arg treatment. Arg enhanced pp70(s6k), cell migration (55% wound coverage), and NO production. In comparison, the branched-chain amino acid leucine (Leu) activated pp70(s6k), was a weaker stimulator of migration (23% coverage), and did not increase NO. A total of 25 micromol/L DETA-NONOate (DETA/NO) did not significantly enhance phosphorylation of p70(s6k) but enhanced the rate of cell migration by approximately 25%. Wound coverage with Leu plus DETA/NO (25 micromol/L) was greater than coverage with DETA/NO alone (P < 0.01). These and our previous studies lead to a model in which Arg must stimulate both pp70(s6k) (in the nucleus) and NO release to enhance intestinal epithelial cell migration, which may be relevant to diseases that involve intestinal villous injury.
Collapse
Affiliation(s)
- J Marc Rhoads
- Department of Pediatrics, University of Texas Health Science Center, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
40
|
Lagranha CJ, Doi SQ, Pithon-Curi TC, Curi R, Sellitti DF. Glutamine enhances glucose-induced mesangial cell proliferation. Amino Acids 2007; 34:683-5. [PMID: 18060527 DOI: 10.1007/s00726-007-0002-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 11/09/2007] [Indexed: 11/30/2022]
Abstract
The proliferation of mesangial cells (MC) in the presence of glutamine (0-20 mM) was determined in both low (5 mM) and high (25 mM) glucose-containing medium. Glutamine in a high glucose (HG) environment increased cell proliferation in a dose-dependent manner. Inhibition of glutamine:fructose 6-phosphate amidotransferase (GFAT) and of phosphodiesterase significantly reduced glutamine-induced proliferation. Supraphysiologic levels of glutamine increase MC proliferation in a HG milieu via GFAT and cAMP-dependent pathways, suggesting that glutamine could pose a risk for diabetic nephropathy.
Collapse
Affiliation(s)
- Claudia J Lagranha
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 1524, 05508-900 São Paulo, SP, Brazil.
| | | | | | | | | |
Collapse
|
41
|
Phanvijhitsiri K, Musch MW, Ropeleski MJ, Chang EB. Heat induction of heat shock protein 25 requires cellular glutamine in intestinal epithelial cells. Am J Physiol Cell Physiol 2006; 291:C290-9. [PMID: 16554407 DOI: 10.1152/ajpcell.00225.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glutamine is considered a nonessential amino acid; however, it becomes conditionally essential during critical illness when consumption exceeds production. Glutamine may modulate the heat shock/stress response, an important adaptive cellular response for survival. Glutamine increases heat induction of heat shock protein (Hsp) 25 in both intestinal epithelial cells (IEC-18) and mesenchymal NIH/3T3 cells, an effect that is neither glucose nor serum dependent. Neither arginine, histidine, proline, leucine, asparagine, nor tyrosine acts as physiological substitutes for glutamine for heat induction of Hsp25. The lack of effect of these amino acids was not caused by deficient transport, although some amino acids, including glutamate (a major direct metabolite of glutamine), were transported poorly by IEC-18 cells. Glutamate uptake could be augmented in a concentration- and time-dependent manner by increasing either media concentration and/or duration of exposure. Under these conditions, glutamate promoted heat induction of Hsp25, albeit not as efficiently as glutamine. Further evidence for the role of glutamine conversion to glutamate was obtained with the glutaminase inhibitor 6-diazo-5-oxo-l-norleucine (DON), which inhibited the effect of glutamine on heat-induced Hsp25. DON inhibited phosphate-dependent glutaminase by 75% after 3 h, decreasing cell glutamate. Increased glutamine/glutamate conversion to glutathione was not involved, since the glutathione synthesis inhibitor, buthionine sulfoximine, did not block glutamine’s effect on heat induction of Hsp25. A large drop in ATP levels did not appear to account for the diminished Hsp25 induction during glutamine deficiency. In summary, glutamine is an important amino acid, and its requirement for heat-induced Hsp25 supports a role for glutamine supplementation to optimize cellular responses to pathophysiological stress.
Collapse
Affiliation(s)
- Kittiporn Phanvijhitsiri
- Department of Biomedical Sciences (Committee on Molecular Metabolism and Nutrition), Martin Boyer Laboratories, Univ. of Chicago IBD Research Center, 5841 S. Maryland Ave., MC6084, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
42
|
Oates PS, West AR. Heme in intestinal epithelial cell turnover, differentiation, detoxification, inflammation, carcinogenesis, absorption and motility. World J Gastroenterol 2006; 12:4281-95. [PMID: 16865768 PMCID: PMC4087737 DOI: 10.3748/wjg.v12.i27.4281] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract is lined by a simple epithelium that undergoes constant renewal involving cell division, differentiation and cell death. In addition, the epithelial lining separates the hostile processes of digestion and absorption that occur in the intestinal lumen from the aseptic environment of the internal milieu by defensive mechanisms that protect the epithelium from being breached. Central to these defensive processes is the synthesis of heme and its catabolism by heme oxygenase (HO). Dietary heme is also an important source of iron for the body which is taken up intact by the enterocyte. This review describes the recent literature on the diverse properties of heme/HO in the intestine tract. The roles of heme/HO in the regulation of the cell cycle/apoptosis, detoxification of xenobiotics, oxidative stress, inflammation, development of colon cancer, heme-iron absorption and intestinal motility are specifically examined.
Collapse
|
43
|
Thébault S, Deniel N, Marion R, Charlionet R, Tron F, Cosquer D, Leprince J, Vaudry H, Ducrotté P, Déchelotte P. Proteomic analysis of glutamine-treated human intestinal epithelial HCT-8 cells under basal and inflammatory conditions. Proteomics 2006; 6:3926-37. [PMID: 16739128 DOI: 10.1002/pmic.200500714] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glutamine (Gln) promotes intestinal growth and maintains gut structure and function, especially in situations of injury and during inflammation. Several mechanisms could contribute to Gln protective effects on gut. Proteomics enable us to characterize differentially expressed proteins in tissues in response to modifications of the biological or nutritional environment. Gln effects on the human intestinal epithelial HCT-8 cell line proteome were assessed under basal and proinflammatory conditions. The 2-DE gels were obtained and compared. Proteins were identified by MS and using databases. About 1200 spots were detected in both 2- and 10-mM Gln concentrations. Under basal conditions, 24 proteins were differentially expressed in response to Gln. Half of these proteins were implicated in protein biosynthesis or proteolysis and 20% in membrane trafficking. Under proinflammatory conditions, 27 proteins were up- or down-regulated by Gln 10 mM. From these proteins, 40% were involved in protein biosynthesis or proteolysis, 16% in membrane trafficking, 8% in cell cycle and apoptosis mechanisms and 8% in nucleic acid metabolism. This study provides the first holistic picture of proteome modulation by Gln in a human enterocytic cell line under basal and proinflammatory conditions, and supports further evaluation of nutritional modulation of intestinal proteome in humans.
Collapse
Affiliation(s)
- Sandrine Thébault
- Groupe ADEN EA3234, Université de Rouen, IFR 23, 22 Boulevard Gambetta, 76183 Rouen, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Häberle J, Görg B, Rutsch F, Schmidt E, Toutain A, Benoist JF, Gelot A, Suc AL, Höhne W, Schliess F, Häussinger D, Koch HG. Congenital glutamine deficiency with glutamine synthetase mutations. N Engl J Med 2005; 353:1926-33. [PMID: 16267323 DOI: 10.1056/nejmoa050456] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glutamine synthetase plays a major role in ammonia detoxification, interorgan nitrogen flux, acid-base homeostasis, and cell signaling. We report on two unrelated newborns who had congenital human glutamine synthetase deficiency with severe brain malformations resulting in multiorgan failure and neonatal death. Glutamine was largely absent from their serum, urine, and cerebrospinal fluid. Each infant had a homozygous mutation in the glutamine synthetase gene (R324C and R341C). Studies that used immortalized lymphocytes expressing R324C glutamine synthetase (R324C-GS) and COS7 cells expressing R341C-GS suggest that these mutations are associated with reduced glutamine synthetase activity.
Collapse
MESH Headings
- Amino Acid Metabolism, Inborn Errors/genetics
- Amino Acid Metabolism, Inborn Errors/metabolism
- Amino Acid Metabolism, Inborn Errors/pathology
- Brain/pathology
- Brain Diseases, Metabolic, Inborn/genetics
- Brain Diseases, Metabolic, Inborn/metabolism
- Brain Diseases, Metabolic, Inborn/pathology
- DNA Mutational Analysis
- Fatal Outcome
- Female
- Glutamate-Ammonia Ligase/deficiency
- Glutamate-Ammonia Ligase/genetics
- Glutamate-Ammonia Ligase/metabolism
- Glutamine/deficiency
- Humans
- Infant, Newborn
- Male
- Point Mutation
Collapse
Affiliation(s)
- Johannes Häberle
- Universitätsklinikum Münster, Klinik und Poliklinik für Kinderheilkunde und Jugendmedizin, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bungard CI, McGivan JD. Glutamine availability up-regulates expression of the amino acid transporter protein ASCT2 in HepG2 cells and stimulates the ASCT2 promoter. Biochem J 2005; 382:27-32. [PMID: 15175006 PMCID: PMC1133911 DOI: 10.1042/bj20040487] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2004] [Revised: 05/25/2004] [Accepted: 06/03/2004] [Indexed: 01/19/2023]
Abstract
Glutamine transport into the human hepatoma cell line HepG2 is catalysed primarily by an ASCT2-type transporter identical in sequence with that cloned previously from JAR cells. An antibody raised against the C-terminus of the ASCT2 protein was shown to recognize ASCT2 on Western blots. Using this antibody, it was found that variation in cell growth rate did not affect ASCT2 expression, but both growth rate and ASCT2 expression were significantly reduced by glutamine deprivation. Expression of a number of other proteins was shown to be unaffected under these conditions. The sequence of the 5'-flanking region of the ASCT2 gene was derived from the human genome database. A 907 bp fragment of this sequence was directionally ligated into a luciferase reporter vector and was shown to exhibit promoter activity when transfected into HepG2 cells. Promoter activity was greatly reduced when transfection was performed in glutamine-free medium and was restored when glutamine was added post-transfection. The absence of other essential amino acids did not affect promoter activity, and glutamine deprivation did not affect the MCT1 (monocarboxylate transporter 1) promoter. These results indicate that both ASCT2 promoter activity and ASCT2 protein expression in these cells are dependent on glutamine availability.
Collapse
Affiliation(s)
- Claire I Bungard
- Department of Biochemistry, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| | | |
Collapse
|
46
|
Blanc MC, Moinard C, Béziel A, Darquy S, Cynober L, De Bandt JP. Arginine and glutamine availability and macrophage functions in the obese insulin-resistant Zucker rat. J Cell Physiol 2005; 202:153-9. [PMID: 15389544 DOI: 10.1002/jcp.20092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increased susceptibility to infections in obese patients may be related to decreased availability of arginine and glutamine, which may affect immune cell functions. Our aim was to evaluate the in vitro effects of these amino acids on the function of macrophages from obese insulin-resistant Zucker rats. Macrophages, isolated from male Zucker obese or lean rats by peritoneal lavage, were incubated in Dulbecco's modified Eagle medium (DMEM) without arginine or glutamine. Arginine or glutamine was added to the medium at increasing final concentrations (0, 0.25, 0.5, 1 or 2 mM). After stimulation by lipopolysaccharide (LPS) from E. coli (40 microg/ml), productions of tumour necrosis factor alpha (TNFalpha) and of nitric oxide (NO) were measured after 3 or 48 h incubation, respectively. NO production, lower in macrophages from obese rats, decreased in macrophages from lean rats (0 mM: 2,423 +/- 1,174 vs. 2 mM: 198 +/- 31 microM/mg protein/24 h; P < 0.05), but not in those from obese rats, when glutamine was added. TNFalpha production, lower in macrophages from obese rats, was inversely correlated with glutamine concentration. In the presence of arginine, NO production was constantly higher in macrophages from obese rats. It peaked at 0.5 mM arginine and decreased thereafter in both groups. TNFalpha production in macrophages from lean rats was unaffected by arginine, but decreased in macrophages from obese rats (0 mM: 1920 +/- 450 vs. 2 mM: 810 +/- 90 microM/mg protein/3 h; P < 0.05). These results suggest that abnormalities in cell signalling or in arginine and glutamine metabolism in macrophages of obese rats, resulting in decreased TNFalpha production and increased NO release, may contribute to increased susceptibility to infection in insulin-resistant states.
Collapse
Affiliation(s)
- Marie-Céline Blanc
- Laboratoire de Biologie de la Nutrition, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris 5-René Descartes, Paris, France
| | | | | | | | | | | |
Collapse
|
47
|
Paquette JC, Guérin PJ, Gauthier ER. Rapid induction of the intrinsic apoptotic pathway byL-glutamine starvation. J Cell Physiol 2004; 202:912-21. [PMID: 15389638 DOI: 10.1002/jcp.20194] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
While the amino acid L-glutamine is known to play a role in the survival of several cell types, the underlying molecular mechanisms are still poorly defined. We show in this report that L-glutamine starvation rapidly triggered apoptosis in Sp2/0-Ag14 hybridoma cells. This process involved the activation of both caspases-9 and -3, suggesting that L-glutamine deprivation initiated an intrinsic apoptotic pathway in Sp2/0-Ag14 cells. Supporting this idea, the cytosolic release of the mitochondrial proteins SMAC/DIABLO and cytochrome c (Cyt c) was observed, with an initial limited leakage occurring during the first 30 min of L-glutamine deprivation, followed by a greater release after 60 min. The latter occurred simultaneously with the translocation of the pro-apoptotic protein Bax to the mitochondria. Finally, a decline in XIAP levels and the activation of caspases-3 and -9 were observed. Thus, L-glutamine deprivation of Sp2/0-Ag14 cells rapidly triggers intracellular events, which target the mitochondria, leading to the cytosolic release of apoptogenic factors, the activation of caspases-9 and -3, and the commitment to the death program. This work introduces the Sp2/0Ag14 hybridoma as a unique model for the study of the molecular events underlying the pro-survival function of L-glutamine.
Collapse
Affiliation(s)
- Julie C Paquette
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | | | | |
Collapse
|
48
|
Deschênes C, Alvarez L, Lizotte ME, Vézina A, Rivard N. The nucleocytoplasmic shuttling of E2F4 is involved in the regulation of human intestinal epithelial cell proliferation and differentiation. J Cell Physiol 2004; 199:262-73. [PMID: 15040009 DOI: 10.1002/jcp.10455] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The specific mechanisms controlling the transition from proliferation to terminal differentiation in human intestinal epithelial cells (HIEC) remain largely undefined. Herein, we analyzed the expression and localization of Rb and E2F proteins in well-established normal intestinal epithelial cell models which allow for the re-enactment of the crypt-villus axis in vitro as well as in intact epithelium and in colon cancer cells. We report that (1) expression of E2F1 is down-regulated while E2F4 protein is sequestered in the cytoplasm during G(0) arrest associated with serum deprivation, confluency, and terminal differentiation of intestinal cells; (2) concurrently, there is an accumulation of the hypophosphorylated form of the pocket proteins into the nucleus with an increased association of E2F4 with pRb and p130; (3) cells which expressed high levels of nuclear E2F4 are all positive for Ki67 staining in human fetal intestine; (4) activation of HIEC crypt cells by growth factors leads to an increase in the nuclear localization of E2F4 which may be attributable to a decrease in the serine/threonine phosphorylation of this transcription factor; (5) inhibition of p38 MAP kinase with alpha/beta inhibitor SB203580 induces E2F4 translocation into the nucleus and its transcriptional activity. In conclusion, our data suggest a key role for E2F4 in proliferation of human intestinal crypt cells and that its cytoplasmic retention as well as its sequestration by Rb proteins may represent a critical step in initiating cell-cycle exit.
Collapse
Affiliation(s)
- Claude Deschênes
- CIHR Group on Functional Development and Physiopathology of the Digestive Tract, Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Manipulation de l’anabolisme au cours des états d’agression. NUTR CLIN METAB 2004. [DOI: 10.1016/j.nupar.2004.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|