1
|
Nakamura A, Matsumoto M. Role of polyamines in intestinal mucosal barrier function. Semin Immunopathol 2025; 47:9. [PMID: 39836273 PMCID: PMC11750915 DOI: 10.1007/s00281-024-01035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
The intestinal epithelium is a rapidly self-renewing tissue; the rapid turnover prevents the invasion of pathogens and harmful components from the intestinal lumen, preventing inflammation and infectious diseases. Intestinal epithelial barrier function depends on the epithelial cell proliferation and junctions, as well as the state of the immune system in the lamina propria. Polyamines, particularly putrescine, spermidine, and spermine, are essential for many cell functions and play a crucial role in mammalian cellular homeostasis, such as that of cell growth, proliferation, differentiation, and maintenance, through multiple biological processes, including translation, transcription, and autophagy. Although the vital role of polyamines in normal intestinal epithelial cell growth and barrier function has been known since the 1980s, recent studies have provided new insights into this topic at the molecular level, such as eukaryotic initiation factor-5A hypusination and autophagy, with rapid advances in polyamine biology in normal cells using biological technologies. This review summarizes recent advances in our understanding of the role of polyamines in regulating normal, non-cancerous, intestinal epithelial barrier function, with a particular focus on intestinal epithelial renewal, cell junctions, and immune cell differentiation in the lamina propria.
Collapse
Affiliation(s)
- Atsuo Nakamura
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd, 20-1 Hirai, Hinode-Machi, Nishitama-Gun, Tokyo, 190-0182, Japan
| | - Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd, 20-1 Hirai, Hinode-Machi, Nishitama-Gun, Tokyo, 190-0182, Japan.
| |
Collapse
|
2
|
Kapur N, Alam MA, Hassan SA, Patel PH, Wempe LA, Bhogoju S, Goretsky T, Kim JH, Herzog J, Ge Y, Awuah SG, Byndloss M, Baumler AJ, Zadeh MM, Sartor RB, Barrett T. Enhanced mucosal mitochondrial function corrects dysbiosis and OXPHOS metabolism in IBD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.584471. [PMID: 38559035 PMCID: PMC10979996 DOI: 10.1101/2024.03.14.584471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background Mitochondrial (Mito) dysfunction in IBD reduces mucosal O2 consumption and increases O2 delivery to the microbiome. Increased enteric O2 promotes blooms of facultative anaerobes (eg. Proteobacteria ) and restricts obligate anaerobes (eg. Firmicutes ). Dysbiotic metabolites negatively affect host metabolism and immunity. Our novel compound (AuPhos) upregulates intestinal epithelial cell (IEC) mito function, attenuates colitis and corrects dysbiosis in humanized Il10-/- mice. We posit that AuPhos corrects IBD-associated dysbiotic metabolism. Methods Primary effect of AuPhos on mucosal Mito respiration and healing process was studied in ex vivo treated human colonic biopsies and piroxicam-accelerated (Px) Il10-/- mice. Secondary effect on microbiome was tested in DSS-colitis WT B6 and germ-free 129.SvEv WT or Il10-/- mice reconstituted with human IBD stool (Hu- Il10-/- ). Mice were treated orally with AuPhos (10- or 25- mg/kg; q3d) or vehicle, stool samples collected for fecal lipocalin-2 (f-LCN2) assay and microbiome analyses using 16S rRNA sequencing. AuPhos effect on microbial metabolites was determined using untargeted global metabolomics. AuPhos-induced hypoxia in IECs was assessed by Hypoxyprobe-1 staining in sections from pimonidazole HCl-infused DSS-mice. Effect of AuPhos on enteric oxygenation was assessed by E. coli Nissle 1917 WT (aerobic respiration-proficient) and cytochrome oxidase (cydA) mutant (aerobic respiration-deficient). Results Metagenomic (16S) analysis revealed AuPhos reduced relative abundances of Proteobacteria and increased blooms of Firmicutes in uninflamed B6 WT, DSS-colitis, Hu-WT and Hu- Il10-/- mice. AuPhos also increased hypoxyprobe-1 staining in surface IECs suggesting enhanced O2 utilization. AuPhos-induced anaerobiosis was confirmed by a significant increase in cydA mutant compared to WT (O2-utlizing) E.coli . Ex vivo treatment of human biopsies with AuPhos showed significant increase in Mito mass, and complexes I and IV. Further, gene expression analysis of AuPhos-treated biopsies showed increase in stem cell markers (Lgr4, Lgr5, Lrig1), with concomitant decreases in pro-inflammatory markers (IL1β,MCP1, RankL). Histological investigation of AuPhos-fed Px- Il10-/- mice showed significantly decreased colitis score in AuPhos-treated Px- Il10-/- mice, with decrease in mRNA of pro-inflammatory cytokines and increase in Mito complexes ( ND5 , ATP6 ). AuPhos significantly altered microbial metabolites associated with SCFA synthesis, FAO, TCA cycle, tryptophan and polyamine biosynthesis pathways. AuPhos increased pyruvate, 4-hydroxybutyrate, 2-hydroxyglutarate and succinate, suggesting an upregulation of pyruvate and glutarate pathways of butyrate production. AuPhos reduced IBD-associated primary bile acids (BA) with concomitant increase in secondary BA (SBA). AuPhos treatment significantly decreased acylcarnitines and increased L-carnitine reflective of enhanced FAO. AuPhos increases TCA cycle intermediates and creatine, energy reservoir substrates indicating enhanced OxPHOS. Besides, AuPhos also upregulates tryptophan metabolism, decreases Kynurenine and its derivatives, and increases polyamine biosynthesis pathway (Putresceine and Spermine). Conclusion These findings indicate that AuPhos-enhanced IEC mitochondrial function reduces enteric O2 delivery, which corrects disease-associated metabolomics by restoring short-chain fatty acids, SBA, AA and IEC energy metabolism. Graphical abstract
Collapse
|
3
|
Arenas-Gómez CM, Garcia-Gutierrez E, Escobar JS, Cotter PD. Human gut homeostasis and regeneration: the role of the gut microbiota and its metabolites. Crit Rev Microbiol 2023; 49:764-785. [PMID: 36369718 DOI: 10.1080/1040841x.2022.2142088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 08/18/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022]
Abstract
The healthy human gut is a balanced ecosystem where host cells and representatives of the gut microbiota interact and communicate in a bidirectional manner at the gut epithelium. As a result of these interactions, many local and systemic processes necessary for host functionality, and ultimately health, take place. Impairment of the integrity of the gut epithelium diminishes its ability to act as an effective gut barrier, can contribute to conditions associated to inflammation processes and can have other negative consequences. Pathogens and pathobionts have been linked with damage of the integrity of the gut epithelium, but other components of the gut microbiota and some of their metabolites can contribute to its repair and regeneration. Here, we review what is known about the effect of bacterial metabolites on the gut epithelium and, more specifically, on the regulation of repair by intestinal stem cells and the regulation of the immune system in the gut. Additionally, we explore the potential therapeutic use of targeted modulation of the gut microbiota to maintain and improve gut homeostasis as a mean to improve health outcomes.
Collapse
Affiliation(s)
- Claudia Marcela Arenas-Gómez
- Vidarium-Nutrition, Health and Wellness Research Center, Grupo Empresarial Nutresa, Medellin, Colombia
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia
| | - Enriqueta Garcia-Gutierrez
- Teagasc Food Research Centre Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- VistaMilk SFI Research Centre, Moorepark, Fermoy, Ireland
| | - Juan S Escobar
- Vidarium-Nutrition, Health and Wellness Research Center, Grupo Empresarial Nutresa, Medellin, Colombia
| | - Paul D Cotter
- Teagasc Food Research Centre Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- VistaMilk SFI Research Centre, Moorepark, Fermoy, Ireland
| |
Collapse
|
4
|
Hodkovicova N, Halas S, Tosnerova K, Stastny K, Svoboda M. The use of functional amino acids in different categories of pigs - A review. VET MED-CZECH 2023; 68:299-312. [PMID: 37982122 PMCID: PMC10646542 DOI: 10.17221/72/2023-vetmed] [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: 06/29/2023] [Accepted: 08/11/2023] [Indexed: 11/21/2023] Open
Abstract
The present review deals with a particularly important topic: the use of functional amino acids in different categories of pigs. It is especially relevant in the context of the current efforts to reduce the use of antibiotics in pig farming and the search for possible alternatives to replace them. The review is based on the definition that functional amino acids (FAAs) are classified as dispensable amino acids, but with additional biological functions, i.e., not only are they used for protein formation, but they are also involved in regulating essential metabolic pathways to improve health, survival, growth, and development. We describe the mechanism of action of individual FAAs and their potential use in pigs, including glutamate, glutamine, arginine, branched-chain amino acids (i.e., leucine, isoleucine, and valine), tryptophan and glycine. The work is divided into three parts. The first part deals with the FAAs and their role in the overall health of sows and their offspring. The second part describes the use of functional amino acids in piglets after weaning. Part three examines the use of functional amino acids in growing and fattening pigs and their impact on meat quality.
Collapse
Affiliation(s)
- Nikola Hodkovicova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Simon Halas
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovak Republic
| | - Kristina Tosnerova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Kamil Stastny
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Martin Svoboda
- Ruminant and Swine Clinic, University of Veterinary Sciences Brno, Brno, Czech Republic
| |
Collapse
|
5
|
Nakamura A, Kurihara S, Takahashi D, Ohashi W, Nakamura Y, Kimura S, Onuki M, Kume A, Sasazawa Y, Furusawa Y, Obata Y, Fukuda S, Saiki S, Matsumoto M, Hase K. Symbiotic polyamine metabolism regulates epithelial proliferation and macrophage differentiation in the colon. Nat Commun 2021; 12:2105. [PMID: 33833232 PMCID: PMC8032791 DOI: 10.1038/s41467-021-22212-1] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal microbiota-derived metabolites have biological importance for the host. Polyamines, such as putrescine and spermidine, are produced by the intestinal microbiota and regulate multiple biological processes. Increased colonic luminal polyamines promote longevity in mice. However, no direct evidence has shown that microbial polyamines are incorporated into host cells to regulate cellular responses. Here, we show that microbial polyamines reinforce colonic epithelial proliferation and regulate macrophage differentiation. Colonisation by wild-type, but not polyamine biosynthesis-deficient, Escherichia coli in germ-free mice raises intracellular polyamine levels in colonocytes, accelerating epithelial renewal. Commensal bacterium-derived putrescine increases the abundance of anti-inflammatory macrophages in the colon. The bacterial polyamines ameliorate symptoms of dextran sulfate sodium-induced colitis in mice. These effects mainly result from enhanced hypusination of eukaryotic initiation translation factor. We conclude that bacterial putrescine functions as a substrate for symbiotic metabolism and is further absorbed and metabolised by the host, thus helping maintain mucosal homoeostasis in the intestine.
Collapse
Affiliation(s)
- Atsuo Nakamura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
- Dairy Science and Technology Institute, Kyodo Milk Industry Co., Ltd., Hinode-machi, Nishitama-gun, Tokyo, Japan
| | - Shin Kurihara
- Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
- Faculty of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
| | - Wakana Ohashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
| | - Yutaka Nakamura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
| | - Shunsuke Kimura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Masayoshi Onuki
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
| | - Aiko Kume
- Dairy Science and Technology Institute, Kyodo Milk Industry Co., Ltd., Hinode-machi, Nishitama-gun, Tokyo, Japan
| | - Yukiko Sasazawa
- Department of Neurology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Yukihiro Furusawa
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa, Toyama, Japan
| | - Yuuki Obata
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan
- The Francis Crick Institute, London, UK
| | - Shinji Fukuda
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan
| | - Shinji Saiki
- Department of Neurology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co., Ltd., Hinode-machi, Nishitama-gun, Tokyo, Japan.
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Minato-ku, Tokyo, Japan.
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
6
|
Matsumoto M. Prevention of Atherosclerosis by the Induction of Microbial Polyamine Production in the Intestinal Lumen. Biol Pharm Bull 2020; 43:221-229. [PMID: 32009110 DOI: 10.1248/bpb.b19-00855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Low molecular weight metabolites produced by the intestinal microbiome that have been associated with health and disease as metabolites need to be constantly absorbed from the intestinal lumen and transported to intestinal epithelial cells and blood. Polyamines, especially spermidine and spermine, are bioactive chemicals which promote autophagy and suppress inflammation. The main source of exogenous polyamines is the intestinal lumen, where they are produced by intestinal microbiome. Considering the intestinal microbiome as a manufacturing plant for bioactive substances, we developed a novel hybrid putrescine biosynthesis system strategy, in which the simultaneous intake of Bifidobacterium animalis ssp. lactis LKM512 (Bifal) and arginine (Arg) upregulates the production of the putrescine, a precursor of spermidine and spermine, in the gut by controlling the bacterial metabolism beyond its vast diversity and inter-individual differences. In a clinical trial, healthy individuals with a body mass index near the maximum "healthy" range (25 kg/m3; n = 44) were randomized to consume either normal yogurt containing Bifal and Arg (Bifal + Arg YG) or placebo (normal yogurt) for 12 weeks. The change in reactive hyperemia index determined by EndoPAT from week 0 to 12 in the Bifal + Arg YG group was significantly higher than that in the placebo group, indicating that Bifal + Arg YG intake improved vascular endothelial function. In addition, the concentrations of fecal putrescine and serum spermidine in the Bifal+ Arg YG group were significantly higher than those in the placebo group. These findings suggest that consuming Bifal + Arg YG prevents or reduces atherosclerosis risk by upregulating blood spermidine levels, which subsequently induces autophagy.
Collapse
|
7
|
Gonzalez-Esquerra R, Leeson S. Physiological and metabolic responses of broilers to heat stress - implications for protein and amino acid nutrition. WORLD POULTRY SCI J 2019. [DOI: 10.1079/wps200597] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- R. Gonzalez-Esquerra
- Department of Animal & Poultry Science, University of Guelph, Guelph, ON, N1G, 2W1, Canada
| | - S. Leeson
- Department of Animal & Poultry Science, University of Guelph, Guelph, ON, N1G, 2W1, Canada
| |
Collapse
|
8
|
Metabolism of the neurotoxic amino acid β-N-methylamino-L-alanine in human cell culture models. Toxicon 2019; 168:131-139. [PMID: 31330193 DOI: 10.1016/j.toxicon.2019.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022]
Abstract
Human dietary exposure to the environmental neurotoxin β-N-methylamino-L-alanine (BMAA) has been implicated in an increased risk of developing sporadic neurodegenerative diseases like Alzheimer's and amyotrophic lateral sclerosis. Evidence suggests that humans are exposed to BMAA globally, but very little is known about BMAA metabolism in mammalian systems, let alone in humans. The most plausible, evidence-based mechanisms of BMAA toxicity rely on the metabolic stability of the amino acid and that, following ingestion, it enters the circulatory system unmodified. BMAA crosses from the intestinal lumen into the circulatory system, and the small intestine and liver are the first sites for dietary amino acid metabolism. Both tissues have substantial amino acid metabolic needs, which are largely fulfilled by dietary amino acids. Metabolism of BMAA in these tissues has been largely overlooked, yet is important in gauging the true human exposure risk. Here we investigate the potential for BMAA metabolism by the human liver and small intestine, using in vitro cell systems. Data show that BMAA metabolism via common proteinogenic amino acid metabolic pathways is negligible, and that in the presence of other amino acids cellular uptake of BMAA is substantially reduced. These data suggest that the majority of ingested BMAA remains unmodified following passage through the small intestine and liver. This not only supports oral BMAA exposure as a plausible exposure route to toxic doses of BMAA, but also supports previous notions that protein deficient diets or malnutrition may increase an individual's susceptibility to BMAA absorption and subsequent toxicity.
Collapse
|
9
|
Yang Y, Kumrungsee T, Kuroda M, Yamaguchi S, Kato N. Feeding Aspergillus protease preparation combined with adequate protein diet to rats increases levels of cecum gut-protective amino acids, partially linked to Bifidobacterium and Lactobacillus. Biosci Biotechnol Biochem 2019; 83:1901-1911. [PMID: 31181987 DOI: 10.1080/09168451.2019.1627183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our recent study indicated that dietary Aspergillus oryzae-derived protease preparation (AP), through its enzymatic activity, exerted a bifidogenic effect in rats. We hypothesized that dietary AP links to protein degradation and subsequently elevates gut-protective amino acids (AAs) in rats fed adequate protein diet. In this study, dietary AP markedly increased the relative abundance of Bifidobacterium and Lactobacillus and the levels of free threonine, alanine, proline, taurine, ornithine, phenylalanine, cystine, and γ-aminobutyric acid in the cecum contents of rats fed with an adequate protein diet, but not in those fed with a low-protein diet. The elevated AAs, except ornithine and phenylalanine, potentially have gut-related health benefits. Some of the AP-modulated free AAs appeared to be associated with the relative abundance of Bifidobacterium and Lactobacillus. Thus, AP combined with adequate protein diet is likely to increase the levels of cecum beneficial free AAs, which is partially associated with the relative abundance of the probiotics.
Collapse
Affiliation(s)
- Yongshou Yang
- Graduate School of Integrated Sciences for Life, Hiroshima University , Higashi-Hiroshima , Japan
| | - Thanutchaporn Kumrungsee
- Graduate School of Integrated Sciences for Life, Hiroshima University , Higashi-Hiroshima , Japan
| | | | | | - Norihisa Kato
- Graduate School of Integrated Sciences for Life, Hiroshima University , Higashi-Hiroshima , Japan
| |
Collapse
|
10
|
Does MHC heterozygosity influence microbiota form and function? PLoS One 2019; 14:e0215946. [PMID: 31095603 PMCID: PMC6522005 DOI: 10.1371/journal.pone.0215946] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
MHC molecules are essential for the adaptive immune response, and they are the most polymorphic genetic loci in vertebrates. Extreme genetic variation at these loci is paradoxical given their central importance to host health. Classic models of MHC gene evolution center on antagonistic host-pathogen interactions to promote gene diversification and allelic diversity in host populations. However, all multicellular organisms are persistently colonized by their microbiota that perform essential metabolic functions for their host and protect from infection. Here, we provide data to support the hypothesis that MHC heterozygote advantage (a main force of selection thought to drive MHC gene evolution), may operate by enhancing fitness advantages conferred by the host’s microbiome. We utilized fecal 16S rRNA gene sequences and their predicted metagenome datasets collected from multiple MHC congenic homozygote and heterozygote mouse strains to describe the influence of MHC heterozygosity on microbiome form and function. We find that in contrast to homozygosity at MHC loci, MHC heterozygosity promotes functional diversification of the microbiome, enhances microbial network connectivity, and results in enrichment for a variety of microbial functions that are positively associated with host fitness. We demonstrate that taxonomic and functional diversity of the microbiome is positively correlated in MHC heterozygote but not homozygote animals, suggesting that heterozygote microbiomes are more functionally adaptive under similar environmental conditions than homozygote microbiomes. Our data complement previous observations on the role of MHC polymorphism in sculpting microbiota composition, but also provide functional insights into how MHC heterozygosity may enhance host health by modulating microbiome form and function. We also provide evidence to support that MHC heterozygosity limits functional redundancy among commensal microbes and may enhance the metabolic versatility of their microbiome. Results from our analyses yield multiple testable predictions regarding the role of MHC heterozygosity on the microbiome that will help guide future research in the area of MHC-microbiome interactions.
Collapse
|
11
|
Xiong X, Tan B, Song M, Ji P, Kim K, Yin Y, Liu Y. Nutritional Intervention for the Intestinal Development and Health of Weaned Pigs. Front Vet Sci 2019; 6:46. [PMID: 30847348 PMCID: PMC6393345 DOI: 10.3389/fvets.2019.00046] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/04/2019] [Indexed: 01/20/2023] Open
Abstract
Weaning imposes simultaneous stress, resulting in reduced feed intake, and growth rate, and increased morbidity and mortality of weaned pigs. Weaning impairs the intestinal integrity, disturbs digestive and absorptive capacity, and increases the intestinal oxidative stress, and susceptibility of diseases in piglets. The improvement of intestinal development and health is critically important for enhancing nutrient digestibility capacity and disease resistance of weaned pigs, therefore, increasing their survival rate at this most vulnerable stage, and overall productive performance during later stages. A healthy gut may include but not limited several important features: a healthy proliferation of intestinal epithelial cells, an integrated gut barrier function, a preferable or balanced gut microbiota, and a well-developed intestinal mucosa immunity. Burgeoning evidence suggested nutritional intervention are one of promising measures to enhance intestinal health of weaned pigs, although the exact protective mechanisms may vary and are still not completely understood. Previous research indicated that functional amino acids, such as arginine, cysteine, glutamine, or glutamate, may enhance intestinal mucosa immunity (i.e., increased sIgA secretion), reduce oxidative damage, stimulate proliferation of enterocytes, and enhance gut barrier function (i.e., enhanced expression of tight junction protein) of weaned pigs. A number of feed additives are marketed to assist in boosting intestinal immunity and regulating gut microbiota, therefore, reducing the negative impacts of weaning, and other environmental challenges on piglets. The promising results have been demonstrated in antimicrobial peptides, clays, direct-fed microbials, micro-minerals, milk components, oligosaccharides, organic acids, phytochemicals, and many other feed additives. This review summarizes our current understanding of nutritional intervention on intestinal health and development of weaned pigs and the importance of mechanistic studies focusing on this research area.
Collapse
Affiliation(s)
- Xia Xiong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Bie Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, South Korea
| | - Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Kwangwook Kim
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| |
Collapse
|
12
|
Gong J, Zhang Z, Zhang X, Chen F, Tan Y, Li H, Jiang J, Zhang J. Effects and possible mechanisms of Alpinia officinarum ethanol extract on indomethacin-induced gastric injury in rats. PHARMACEUTICAL BIOLOGY 2018; 56:294-301. [PMID: 29781354 PMCID: PMC6130516 DOI: 10.1080/13880209.2018.1450426] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
CONTEXT Alpinia officinarum Hance (Zingiberoside) has a long history in treating gastrointestinal diseases, but its mechanisms of action are not yet known. OBJECTIVE To investigate the effects and underlying mechanisms of the ethanol extract of A. officinarum rhizomes in an indomethacin-induced gastric injury rat model. MATERIAL AND METHODS Indomethacin (0.3 g/kg) was orally administered to Sprague-Dawley rats to induce gastric damage; after 7 h, the rats were treated with 0.03, 0.09, or 0.18 g/kg of the plant extract, galangin (0.2 g/kg), or bismuth potassium citrate (0.08 g/kg), once a day for 6 days. Rats in the control group received an equivalent volume of vehicle solution for 6 days. Gastric damage was evaluated by gross ulcer and histological indexes. Cyclooxygenase and non-cyclooxygenase pathway proteins were quantified by western blotting and ELISA. RESULTS Alpinia officinarum extract ameliorated gastric injury in a dose-dependent manner, and 0.18 g/kg dose exhibited the best performance by reducing the gross ulcer (from 20.23 ± 1.38 to 1.66 ± 0.37) and histological (from 4.67 ± 1.03 to 0.33 ± 0.51) indexes, decreasing serum TNF-α level (14.17%), increasing serum VEGF level (1.58 times), increasing cyclooxygenase-1 level (1.25 times, p < 0.001) in the gastric mucosa, and reversing indomethacin-induced changes in the expression of non-cyclooxygenase pathway proteins (p < 0.05). Galangin was less effective as an antiulcer agent than the whole extract, indicating that other components also contributed to the protective effect. CONCLUSIONS Alpinia officinarum extract and galangin exert antiulcer effects through cyclooxygenase and non-cyclooxygenase pathways validating use of galangin as a treatment for gastric damage.
Collapse
Affiliation(s)
- Jingwen Gong
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
| | - Zhong Zhang
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
| | - Xuguang Zhang
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
| | - Feng Chen
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
| | - Yinfeng Tan
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
| | - Hailong Li
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
| | - Jie Jiang
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
- Jie JiangHainan Medical University, 3 Xueyuan Road, Haikou571199, China
| | - Junqing Zhang
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, Hainan Medical University, Haikou, China
- CONTACT Junqing Zhang
| |
Collapse
|
13
|
Wakita Y, Shimomura Y, Kitada Y, Yamamoto H, Ohashi Y, Matsumoto M. Taxonomic classification for microbiome analysis, which correlates well with the metabolite milieu of the gut. BMC Microbiol 2018; 18:188. [PMID: 30445918 PMCID: PMC6240276 DOI: 10.1186/s12866-018-1311-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/10/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND 16S rRNA gene amplicon sequencing analysis (16S amplicon sequencing) has provided considerable information regarding the ecology of the intestinal microbiome. Recently, metabolomics has been used for investigating the crosstalk between the intestinal microbiome and the host via metabolites. In the present study, we determined the accuracy with which 16S rRNA gene data at different classification levels correspond to the metabolome data for an in-depth understanding of the intestinal environment. RESULTS Over 200 metabolites were identified using capillary electrophoresis and time-of-flight mass spectrometry (CE-TOFMS)-based metabolomics in the feces of antibiotic-treated and untreated mice. 16S amplicon sequencing, followed by principal component analysis (PCA) of the intestinal microbiome at each taxonomic rank, revealed differences between the antibiotic-treated and untreated groups in the first principal component in the family-, genus, and species-level analyses. These differences were similar to those observed in the PCA of the metabolome. Furthermore, a strong correlation between principal component (PC) scores of the metabolome and microbiome was observed in family-, genus-, and species-level analyses. CONCLUSIONS Lower taxonomic ranks such as family, genus, or species are preferable for 16S amplicon sequencing to investigate the correlation between the microbiome and metabolome. The correlation of PC scores between the microbiome and metabolome at lower taxonomic levels yield a simple method of integrating different "-omics" data, which provides insights regarding crosstalk between the intestinal microbiome and the host.
Collapse
Affiliation(s)
- Yoshihisa Wakita
- Frontier Laboratories for Value Creation, Sapporo Holdings Ltd., Yaizu, Shizuoka, 425-0013, Japan
| | - Yumi Shimomura
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd., Hinode-machi, Tokyo, 190-0182, Japan
| | - Yusuke Kitada
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd., Hinode-machi, Tokyo, 190-0182, Japan
| | - Hiroyuki Yamamoto
- Human Metabolome Technologies, Inc., Tsuruoka, Yamagata, 997-0052, Japan
| | - Yoshiaki Ohashi
- Human Metabolome Technologies, Inc., Tsuruoka, Yamagata, 997-0052, Japan
| | - Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd., Hinode-machi, Tokyo, 190-0182, Japan.
| |
Collapse
|
14
|
Gerner EW, Bruckheimer E, Cohen A. Cancer pharmacoprevention: Targeting polyamine metabolism to manage risk factors for colon cancer. J Biol Chem 2018; 293:18770-18778. [PMID: 30355737 DOI: 10.1074/jbc.tm118.003343] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer is a set of diseases characterized by uncontrolled cell growth. In certain cancers of the gastrointestinal tract, the adenomatous polyposis coli (APC) tumor suppressor gene is altered in either germline or somatic cells and causes formation of risk factors, such as benign colonic or intestinal neoplasia, which can progress to invasive cancer. APC is a key component of the WNT pathway, contributing to normal GI tract development, and APC alteration results in dysregulation of the pathway for production of polyamines, which are ubiquitous cations essential for cell growth. Studies with mice have identified nonsteroidal anti-inflammatory drugs (NSAIDs) and difluoromethylornithine (DFMO), an inhibitor of polyamine synthesis, as potent inhibitors of colon carcinogenesis. Moreover, gene expression profiling has uncovered that NSAIDs activate polyamine catabolism and export. Several DFMO-NSAID combination strategies are effective and safe methods for reducing risk factors in clinical trials with patients having genetic or sporadic risk of colon cancer. These strategies affect cancer stem cells, inflammation, immune surveillance, and the microbiome. Pharmacotherapies consisting of drug combinations targeting the polyamine pathway provide a complementary approach to surgery and cytotoxic cancer treatments for treating patients with cancer risk factors. In this Minireview, we discuss the role of polyamines in colon cancer and highlight the mechanisms of select pharmacoprevention agents to delay or prevent carcinogenesis in humans.
Collapse
Affiliation(s)
- Eugene W Gerner
- From Cancer Prevention Pharmaceuticals, Tucson, Arizona 85718 and .,the Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona 85711
| | | | - Alfred Cohen
- From Cancer Prevention Pharmaceuticals, Tucson, Arizona 85718 and
| |
Collapse
|
15
|
Sugiyama Y, Nara M, Sakanaka M, Gotoh A, Kitakata A, Okuda S, Kurihara S. Comprehensive analysis of polyamine transport and biosynthesis in the dominant human gut bacteria: Potential presence of novel polyamine metabolism and transport genes. Int J Biochem Cell Biol 2017; 93:52-61. [PMID: 29102547 DOI: 10.1016/j.biocel.2017.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/23/2017] [Accepted: 10/31/2017] [Indexed: 10/18/2022]
Abstract
Recent studies have reported that polyamines in the colonic lumen might affect animal health and these polyamines are thought to be produced by gut bacteria. In the present study, we measured the concentrations of three polyamines (putrescine, spermidine, and spermine) in cells and culture supernatants of 32 dominant human gut bacterial species in their growing and stationary phases. Combining polyamine concentration analysis in culture supernatant and cells with available genomic information showed that novel polyamine biosynthetic proteins and transporters were present in dominant human gut bacteria. Based on these findings, we suggested strategies for optimizing polyamine concentrations in the human colonic lumen via regulation of genes responsible for polyamine biosynthesis and transport in the dominant human gut bacteria.
Collapse
Affiliation(s)
- Yuta Sugiyama
- Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Misaki Nara
- Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | | | - Aina Gotoh
- Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Aya Kitakata
- Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Shujiro Okuda
- Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Shin Kurihara
- Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan.
| |
Collapse
|
16
|
Asquith M, Davin S, Stauffer P, Michell C, Janowitz C, Lin P, Ensign-Lewis J, Kinchen JM, Koop DR, Rosenbaum JT. Intestinal Metabolites Are Profoundly Altered in the Context of HLA-B27 Expression and Functionally Modulate Disease in a Rat Model of Spondyloarthritis. Arthritis Rheumatol 2017; 69:1984-1995. [PMID: 28622455 PMCID: PMC5623151 DOI: 10.1002/art.40183] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 06/13/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE HLA-B27-associated spondyloarthritides are associated with an altered intestinal microbiota and bowel inflammation. We undertook this study to identify HLA-B27-dependent changes in both host and microbial metabolites in the HLA-B27/β2 -microglobulin (β2 m)-transgenic rat and to determine whether microbiota-derived metabolites could impact disease in this major model of spondyloarthritis. METHODS Cecal contents were collected from Fischer 344 33-3 HLA-B27/β2 m-transgenic rats and wild-type controls at 6 weeks (before disease) and 16 weeks (with active bowel inflammation). Metabolomic profiling was performed by high-throughput gas and liquid chromatography-based mass spectrometry. HLA-B27/β2 m-transgenic rats were treated with the microbial metabolites propionate or butyrate in drinking water for 10 weeks, and disease activity was subsequently assessed. RESULTS Our screen identified 582 metabolites, of which more than half were significantly altered by HLA-B27 expression at 16 weeks. Both microbial and host metabolites were altered, with multiple pathways affected, including those for amino acid, carbohydrate, xenobiotic, and medium-chain fatty acid metabolism. Differences were even observed at 6 weeks, with up-regulation of histidine, tyrosine, spermidine, N-acetylmuramate, and glycerate in HLA-B27/β2 m-transgenic rats. Administration of the short-chain fatty acid propionate significantly attenuated HLA-B27-associated inflammatory disease, although this was not associated with increased FoxP3+ T cell induction or with altered expression of the immunomodulatory cytokines interleukin-10 (IL-10) or IL-33 or of the tight junction protein zonula occludens 1. HLA-B27 expression was also associated with altered host expression of messenger RNA for the microbial metabolite receptors free fatty acid receptor 2 (FFAR2), FFAR3, and niacin receptor 1. CONCLUSION HLA-B27 expression profoundly impacts the intestinal metabolome, with changes evident in rats even at age 6 weeks. Critically, we demonstrate that a microbial metabolite, propionate, attenuates development of HLA-B27-associated inflammatory disease. These and other microbiota-derived bioactive mediators may provide novel treatment modalities in HLA-B27-associated spondyloarthritides.
Collapse
Affiliation(s)
- Mark Asquith
- Oregon Health and Science University, Division of Arthritis and
Rheumatic Diseases, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
| | - Sean Davin
- Oregon Health and Science University, Division of Arthritis and
Rheumatic Diseases, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
| | - Patrick Stauffer
- Oregon Health and Science University, Division of Arthritis and
Rheumatic Diseases, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
| | - Claire Michell
- Oregon Health and Science University, Division of Arthritis and
Rheumatic Diseases, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
| | - Cathleen Janowitz
- Oregon Health and Science University, Division of Arthritis and
Rheumatic Diseases, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
| | - Phoebe Lin
- Oregon Health and Science University, Casey Eye Institute, Portland,
OR, USA, 97239
| | - Joe Ensign-Lewis
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic
Center Blvd, Philadelphia, PA, USA, 19104
| | | | - Dennis R Koop
- Oregon Health and Science University, Department of Physiology and
Pharmacology, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
| | - James T. Rosenbaum
- Oregon Health and Science University, Division of Arthritis and
Rheumatic Diseases, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
- Oregon Health and Science University, Department of Physiology and
Pharmacology, 3181 SW Sam Jackson Park Rd, Portland, OR, USA, 97239
- Devers Eye Institute, 1040 Nw 22nd Ave, Suite 200, Portland, OR,
USA, 97210
| |
Collapse
|
17
|
Abstract
The human gastrointestinal tract is populated by a diverse, highly mutualistic microbial flora, which is known as the microbiome. Disruptions to the microbiome have been shown to be associated with severe pathologies of the host, including metabolic disease, cancer, and inflammatory bowel disease. Mood and behavior are also susceptible to alterations in the gut microbiota. A particularly striking example of the symbiotic effects of the microbiome is the immune system, whose cells depend critically on a diverse array of microbial metabolites for normal development and behavior. This includes metabolites that are produced by bacteria from dietary components, metabolites that are produced by the host and biochemically modified by gut bacteria, and metabolites that are synthesized de novo by gut microbes. In this review, we highlight the role of the intestinal microbiome in human metabolic and inflammatory diseases and focus in particular on the molecular mechanisms that govern the gut-immune axis.
Collapse
Affiliation(s)
- Thomas Siegmund Postler
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
18
|
Li Z, Wright ADG, Si H, Wang X, Qian W, Zhang Z, Li G. Changes in the rumen microbiome and metabolites reveal the effect of host genetics on hybrid crosses. ENVIRONMENTAL MICROBIOLOGY REPORTS 2016; 8:1016-1023. [PMID: 27717170 DOI: 10.1111/1758-2229.12482] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/29/2016] [Indexed: 06/06/2023]
Abstract
The rumen microbiota plays important roles in nutrient metabolism and absorption of the host. However, it is poorly understood how host genetic variation shapes the community structure of the rumen microbiota and its metabolic phenotype. Here, we used sika deer (Cervus nippon) and elk (Cervus elaphus) to produce the following two types of hybrid offspring: sika deer ♀ × elk ♂ (SEH) and elk ♀ × sika deer ♂ (ESH). Then, we examined the rumen microbiome and metabolites in the parents and their hybrid offspring. The rumen microbiota in the hybrids differed from that in their parents, suggesting a significant effect of host genetics on the rumen microbiome that may have resulted from vertical transmission. The rumen metabolites displayed patterns similar to the structure of the rumen microbiome, with changes in the amounts of volatile fatty acids and metabolites of amino acids. The alanine, arginine, proline and phenylalanine pathways were enriched in the rumen of hybrid animals. The enriched metabolites in the above pathways were positively correlated with the bacteria Prevotella spp., Acetitomaculum spp., Quinella spp., Succinivibrio spp. and Ruminobacter spp. These results suggest that host genetics has a major impact on the rumen microbiome and metabolites in hybrid animals.
Collapse
Affiliation(s)
- Zhipeng Li
- Jilin Provincial Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | | | - Huazhe Si
- Jilin Provincial Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Xiaoxu Wang
- Jilin Provincial Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Wenxi Qian
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhigang Zhang
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| | - Guangyu Li
- Jilin Provincial Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| |
Collapse
|
19
|
Goudarzi M, Mak TD, Jacobs JP, Moon BH, Strawn SJ, Braun J, Brenner DJ, Fornace AJ, Li HH. An Integrated Multi-Omic Approach to Assess Radiation Injury on the Host-Microbiome Axis. Radiat Res 2016; 186:219-34. [PMID: 27512828 PMCID: PMC5304359 DOI: 10.1667/rr14306.1] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Medical responders to radiological and nuclear disasters currently lack sufficient high-throughput and minimally invasive biodosimetry tools to assess exposure and injury in the affected populations. For this reason, we have focused on developing robust radiation exposure biomarkers in easily accessible biofluids such as urine, serum and feces. While we have previously reported on urine and serum biomarkers, here we assessed perturbations in the fecal metabolome resulting from exposure to external X radiation in vivo. The gastrointestinal (GI) system is of particular importance in radiation biodosimetry due to its constant cell renewal and sensitivity to radiation-induced injury. While the clinical GI symptoms such as pain, bloating, nausea, vomiting and diarrhea are manifested after radiation exposure, no reliable bioindicator has been identified for radiation-induced gastrointestinal injuries. To this end, we focused on determining a fecal metabolomic signature in X-ray irradiated mice. There is overwhelming evidence that the gut microbiota play an essential role in gut homeostasis and overall health. Because the fecal metabolome is tightly correlated with the composition and diversity of the microorganism in the gut, we also performed fecal 16S rRNA sequencing analysis to determine the changes in the microbial composition postirradiation. We used in-house bioinformatics tools to integrate the 16S rRNA sequencing and metabolomic data, and to elucidate the gut integrated ecosystem and its deviations from a stable host-microbiome state that result from irradiation. The 16S rRNA sequencing results indicated that radiation caused remarkable alterations of the microbiome in feces at the family level. Increased abundance of common members of Lactobacillaceae and Staphylococcaceae families, and decreased abundances of Lachnospiraceae, Ruminococcaceae and Clostridiaceae families were found after 5 and 12 Gy irradiation. The metabolomic data revealed statistically significant changes in the microbial-derived products such as pipecolic acid, glutaconic acid, urobilinogen and homogentisic acid. In addition, significant changes were detected in bile acids such as taurocholic acid and 12-ketodeoxycholic acid. These changes may be associated with the observed shifts in the abundance of intestinal microbes, such as R. gnavus , which can transform bile acids.
Collapse
Affiliation(s)
- Maryam Goudarzi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Tytus D. Mak
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Jonathan P. Jacobs
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Bo-Hyun Moon
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Steven J. Strawn
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Jonathan Braun
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - David J. Brenner
- Center for Radiological Research, Columbia University, New York, New York
| | - Albert J. Fornace
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Heng-Hong Li
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| |
Collapse
|
20
|
Evageliou NF, Haber M, Vu A, Laetsch TW, Murray J, Gamble LD, Cheng NC, Liu K, Reese M, Corrigan KA, Ziegler DS, Webber H, Hayes CS, Pawel B, Marshall GM, Zhao H, Gilmour SK, Norris MD, Hogarty MD. Polyamine Antagonist Therapies Inhibit Neuroblastoma Initiation and Progression. Clin Cancer Res 2016; 22:4391-404. [PMID: 27012811 DOI: 10.1158/1078-0432.ccr-15-2539] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/15/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Deregulated MYC drives oncogenesis in many tissues yet direct pharmacologic inhibition has proven difficult. MYC coordinately regulates polyamine homeostasis as these essential cations support MYC functions, and drugs that antagonize polyamine sufficiency have synthetic-lethal interactions with MYC Neuroblastoma is a lethal tumor in which the MYC homologue MYCN, and ODC1, the rate-limiting enzyme in polyamine synthesis, are frequently deregulated so we tested optimized polyamine depletion regimens for activity against neuroblastoma. EXPERIMENTAL DESIGN We used complementary transgenic and xenograft-bearing neuroblastoma models to assess polyamine antagonists. We investigated difluoromethylornithine (DFMO; an inhibitor of Odc, the rate-limiting enzyme in polyamine synthesis), SAM486 (an inhibitor of Amd1, the second rate-limiting enzyme), and celecoxib (an inducer of Sat1 and polyamine catabolism) in both the preemptive setting and in the treatment of established tumors. In vitro assays were performed to identify mechanisms of activity. RESULTS An optimized polyamine antagonist regimen using DFMO and SAM486 to inhibit both rate-limiting enzymes in polyamine synthesis potently blocked neuroblastoma initiation in transgenic mice, underscoring the requirement for polyamines in MYC-driven oncogenesis. Furthermore, the combination of DFMO with celecoxib was found to be highly active, alone, and combined with numerous chemotherapy regimens, in regressing established tumors in both models, including tumors harboring highest risk genetic lesions such as MYCN amplification, ALK mutation, and TP53 mutation with multidrug resistance. CONCLUSIONS Given the broad preclinical activity demonstrated by polyamine antagonist regimens across diverse in vivo models, clinical investigation of such approaches in neuroblastoma and potentially other MYC-driven tumors is warranted. Clin Cancer Res; 22(17); 4391-404. ©2016 AACR.
Collapse
Affiliation(s)
- Nicholas F Evageliou
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. Center for Childhood Cancer Research, University of New South Wales, Sydney, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Sydney, Australia
| | - Annette Vu
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Jayne Murray
- Children's Cancer Institute Australia, Sydney, Australia
| | - Laura D Gamble
- Children's Cancer Institute Australia, Sydney, Australia
| | | | - Kangning Liu
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Megan Reese
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kelly A Corrigan
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David S Ziegler
- Children's Cancer Institute Australia, Sydney, Australia. Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia. School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Kensington, Sydney, Australia
| | - Hannah Webber
- Children's Cancer Institute Australia, Sydney, Australia
| | - Candice S Hayes
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Bruce Pawel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Glenn M Marshall
- Children's Cancer Institute Australia, Sydney, Australia. Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Huaqing Zhao
- Department of Biostatistics, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Susan K Gilmour
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Murray D Norris
- Children's Cancer Institute Australia, Sydney, Australia. Center for Childhood Cancer Research, University of New South Wales, Sydney, Australia
| | - Michael D Hogarty
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
21
|
Tsai YH, Lin KL, Huang YP, Hsu YC, Chen CH, Chen Y, Sie MH, Wang GJ, Lee MJ. Suppression of ornithine decarboxylase promotes osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. FEBS Lett 2015; 589:2058-65. [DOI: 10.1016/j.febslet.2015.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 06/08/2015] [Accepted: 06/15/2015] [Indexed: 11/24/2022]
|
22
|
Abstract
Advanced mucosal healing (MH) after intestinal mucosal inflammation coincides with sustained clinical remission and reduced rates of hospitalization and surgical resection, explaining why MH is increasingly considered as a full therapeutic goal and as an endpoint for clinical trials. Intestinal MH is a complex phenomenon viewed as a succession of steps necessary to restore tissue structure and function. These steps include epithelial cell migration and proliferation, cell differentiation, restoration of epithelial barrier functions, and modulation of cell apoptosis. Few clinical studies have evaluated the needs for specific macronutrients and micronutrients and their effects on intestinal MH, most data having been obtained from animal and cell studies. These data suggest that supplementation with specific amino acids including arginine, glutamine, glutamate, threonine, methionine, serine, proline, and the amino acid-derived compounds, polyamines can favorably influence MH. Short-chain fatty acids, which are produced by the microbiota from undigested polysaccharides and protein-derived amino acids, also exert beneficial effects on the process of intestinal MH in experimental models. Regarding supplementation with lipids, although the effects of ω-3 and ω-6 fatty acids remain controversial, endogenous prostaglandin synthesis seems to be necessary for MH. Finally, among micronutrients, several vitamin and mineral deficiencies with different frequencies have been observed in patients with inflammatory bowel diseases and supplementation with some of them (vitamin A, vitamin D3, vitamin C, and zinc) are presumed to favor MH. Future work, including clinical studies, should evaluate the efficiency of supplementation with combination of dietary compounds as adjuvant nutritional intervention for MH of the inflamed intestinal mucosa.
Collapse
|
23
|
Matsumoto M, Ebata T, Hirooka J, Hosoya R, Inoue N, Itami S, Tsuji K, Yaginuma T, Muramatsu K, Nakamura A, Fujita A, Nagakura T. Antipruritic effects of the probiotic strain LKM512 in adults with atopic dermatitis. Ann Allergy Asthma Immunol 2014; 113:209-216.e7. [PMID: 24893766 DOI: 10.1016/j.anai.2014.05.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/23/2014] [Accepted: 05/05/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Evidence suggests that intestinal microbiota play an important role in the pathogenesis of atopic dermatitis (AD) through induction of immunosuppression and immune tolerance; however, the exact underlying mechanism is unclear. Few studies to date have examined the effects of probiotics on adult-type AD. OBJECTIVE To examine the effects of the probiotic Bifidobacterium animalis subsp lactis LKM512 on adult-type AD and the expression of metabolites that are known to be influenced by gut microbiota in fecal samples. METHODS Forty-four patients were randomly assigned to receive LKM512 or a placebo and underwent medical examinations. Fecal microbiota were analyzed with real-time polymerase chain reaction. Metabolomic analysis was conducted to search for antipruritic metabolites produced by intestinal bacteria using feces derived from 3 patients whose itch scores had improved using capillary electrophoresis with time-of-flight mass spectrometry. Antipruritic effects of kynurenic acid were observed using AD-induced NC/Nga mice. RESULTS LKM512 administration alleviated itch in AD patients compared with controls and improved the dermatology-specific quality-of-life scores when compared with the controls. Administration of LKM512 also increased the expression of the antipruritic and antinociceptive metabolite kynurenic acid (KYNA) in patients whose itch score had improved after LKM512 treatment. In mouse experiments, scratching behavior counts tended to be decreased by KYNA injection when compared with no treatment. CONCLUSION LKM512 administration may exert antipruritic effects by increasing KYNA production. LKM512 could therefore be a potentially effective therapeutic candidate for the reduction of pruritus. TRIAL REGISTRATION umin.ac.jp/ctr Identifier: UMIN000005695.
Collapse
Affiliation(s)
- Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co Ltd, Hinode, Tokyo, Japan.
| | - Toshiya Ebata
- Chitofuna Dermatology Clinic, Setagaya, Tokyo, Japan
| | | | | | | | | | | | | | - Koji Muramatsu
- Dairy Science and Technology Institute, Kyodo Milk Industry Co Ltd, Hinode, Tokyo, Japan
| | - Atsuo Nakamura
- Dairy Science and Technology Institute, Kyodo Milk Industry Co Ltd, Hinode, Tokyo, Japan
| | - Ayako Fujita
- Dairy Science and Technology Institute, Kyodo Milk Industry Co Ltd, Hinode, Tokyo, Japan
| | | |
Collapse
|
24
|
Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep 2014; 4:4548. [PMID: 24686447 PMCID: PMC4070089 DOI: 10.1038/srep04548] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/13/2014] [Indexed: 12/12/2022] Open
Abstract
Prevention of quality of life (QOL) deterioration is associated with the inhibition of geriatric diseases and the regulation of brain function. However, no substance is known that prevents the aging of both body and brain. It is known that polyamine concentrations in somatic tissues (including the brain) decrease with increasing age, and polyamine-rich foods enhance longevity in yeast, worms, flies, and mice, and protect flies from age-induced memory impairment. A main source of exogenous polyamines is the intestinal lumen, where they are produced by intestinal bacteria. We found that arginine intake increased the concentration of putrescine in the colon and increased levels of spermidine and spermine in the blood. Mice orally administered with arginine in combination with the probiotic bifidobacteria LKM512 long-term showed suppressed inflammation, improved longevity, and protection from age-induced memory impairment. This study shows that intake of arginine and LKM512 may prevent aging-dependent declines in QOL via the upregulation of polyamines.
Collapse
|
25
|
Lee MJ, Chen Y, Huang YP, Hsu YC, Chiang LH, Chen TY, Wang GJ. Exogenous polyamines promote osteogenic differentiation by reciprocally regulating osteogenic and adipogenic gene expression. J Cell Biochem 2013; 114:2718-28. [DOI: 10.1002/jcb.24620] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 06/17/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Mon-Juan Lee
- Department of Bioscience Technology; Chang Jung Christian University; Tainan; Taiwan
| | | | - Yuan-Pin Huang
- Department of Cosmetics and Fashion Styling; Cheng Shiu University; Kaohsiung; Taiwan
| | - Yi-Chiang Hsu
- Graduate Institute of Medical Sciences; Chang Jung Christian University; Tainan; Taiwan
| | - Lan-Hsin Chiang
- Department of Bioscience Technology; Chang Jung Christian University; Tainan; Taiwan
| | - Tzu-Yu Chen
- Department of Bioscience Technology; Chang Jung Christian University; Tainan; Taiwan
| | | |
Collapse
|
26
|
González-Tenorio R, Fonseca B, Caro I, Fernández-Diez A, Kuri V, Soto S, Mateo J. Changes in biogenic amine levels during storage of Mexican-style soft and Spanish-style dry-ripened sausages with different a(w) values under modified atmosphere. Meat Sci 2013; 94:369-75. [PMID: 23567139 DOI: 10.1016/j.meatsci.2013.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/13/2013] [Accepted: 03/17/2013] [Indexed: 11/15/2022]
Abstract
Two raw sausages were prepared: a soft and a dry-ripened one, both by local traditional and industrial manufacturing practices. Sausages were packaged under a CO2/N2 atmosphere at different targeted activity water (aw) values: 0.96 and 0.92 (soft sausages) and 0.88 and 0.82 (dry-ripened sausages). Sausages were then stored at 5 °C for 42 days or at 12 °C for 240 days (soft and a dry-ripened sausages, respectively). The time-related changes in dominant microbiota, pH and biogenic amine contents during storage were determined. Tyramine was the most abundant biogenic amine in all the sausages. Biogenic amine levels were higher in dry-ripened sausages than in soft sausages at packaging. However, during refrigerated storage soft sausages were fermented and the levels of biogenic amines increased (P<0.05). At the end of storage, traditional soft sausages with 0.96 aw presented comparable levels of biogenic amines to traditional dry-ripened sausages.
Collapse
Affiliation(s)
- Roberto González-Tenorio
- Institute of Agricultural and Livestock Sciences, Universidad Autónoma del Estado de Hidalgo, Av. Universidad s/n Km 1 Rancho Universitario CP. 43600. Tulancingo, Hidalgo, Mexico
| | | | | | | | | | | | | |
Collapse
|
27
|
Balbach ST, Esteves TC, Houghton FD, Siatkowski M, Pfeiffer MJ, Tsurumi C, Kanzler B, Fuellen G, Boiani M. Nuclear reprogramming: kinetics of cell cycle and metabolic progression as determinants of success. PLoS One 2012; 7:e35322. [PMID: 22530006 PMCID: PMC3329427 DOI: 10.1371/journal.pone.0035322] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 03/14/2012] [Indexed: 01/16/2023] Open
Abstract
Establishment of totipotency after somatic cell nuclear transfer (NT) requires not only reprogramming of gene expression, but also conversion of the cell cycle from quiescence to the precisely timed sequence of embryonic cleavage. Inadequate adaptation of the somatic nucleus to the embryonic cell cycle regime may lay the foundation for NT embryo failure and their reported lower cell counts. We combined bright field and fluorescence imaging of histone H2b-GFP expressing mouse embryos, to record cell divisions up to the blastocyst stage. This allowed us to quantitatively analyze cleavage kinetics of cloned embryos and revealed an extended and inconstant duration of the second and third cell cycles compared to fertilized controls generated by intracytoplasmic sperm injection (ICSI). Compared to fertilized embryos, slow and fast cleaving NT embryos presented similar rates of errors in M phase, but were considerably less tolerant to mitotic errors and underwent cleavage arrest. Although NT embryos vary substantially in their speed of cell cycle progression, transcriptome analysis did not detect systematic differences between fast and slow NT embryos. Profiling of amino acid turnover during pre-implantation development revealed that NT embryos consume lower amounts of amino acids, in particular arginine, than fertilized embryos until morula stage. An increased arginine supplementation enhanced development to blastocyst and increased embryo cell numbers. We conclude that a cell cycle delay, which is independent of pluripotency marker reactivation, and metabolic restraints reduce cell counts of NT embryos and impede their development.
Collapse
Affiliation(s)
| | | | - Franchesca Dawn Houghton
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Marcin Siatkowski
- German Center for Neurodegenerative Disorders, DZNE, Rostock, Germany
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, University of Rostock, Rostock, Germany
| | | | - Chizuko Tsurumi
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Benoit Kanzler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, University of Rostock, Rostock, Germany
| | - Michele Boiani
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
- * E-mail:
| |
Collapse
|
28
|
Matsumoto M, Kibe R, Ooga T, Aiba Y, Kurihara S, Sawaki E, Koga Y, Benno Y. Impact of intestinal microbiota on intestinal luminal metabolome. Sci Rep 2012; 2:233. [PMID: 22724057 PMCID: PMC3380406 DOI: 10.1038/srep00233] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 12/23/2011] [Indexed: 12/19/2022] Open
Abstract
Low-molecular-weight metabolites produced by intestinal microbiota play a direct role in health and disease. In this study, we analyzed the colonic luminal metabolome using capillary electrophoresis mass spectrometry with time-of-flight (CE-TOFMS) -a novel technique for analyzing and differentially displaying metabolic profiles- in order to clarify the metabolite profiles in the intestinal lumen. CE-TOFMS identified 179 metabolites from the colonic luminal metabolome and 48 metabolites were present in significantly higher concentrations and/or incidence in the germ-free (GF) mice than in the Ex-GF mice (p < 0.05), 77 metabolites were present in significantly lower concentrations and/or incidence in the GF mice than in the Ex-GF mice (p < 0.05), and 56 metabolites showed no differences in the concentration or incidence between GF and Ex-GF mice. These indicate that intestinal microbiota highly influenced the colonic luminal metabolome and a comprehensive understanding of intestinal luminal metabolome is critical for clarifying host-intestinal bacterial interactions.
Collapse
Affiliation(s)
- Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd., Tokyo 190-0182, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Intestinal gene expression in pigs: effects of reduced feed intake during weaning and potential impact of dietary components. Nutr Res Rev 2011; 24:155-75. [DOI: 10.1017/s0954422411000047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The weaning transition is characterised by morphological, histological and microbial changes, often leading to weaning-associated disorders. These intestinal changes can partly be ascribed to the lack of luminal nutrition arising from the reduced feed intake common in pigs after weaning. It is increasingly becoming clear that changes in the supply with enteral nutrients may have major impacts on intestinal gene expression. Furthermore, the major dietary constituents, i.e. carbohydrates, fatty acids and amino acids, participate in the regulation of intestinal gene expression. However, nutrients may also escape digestion by mammalian enzymes in the upper gastrointestinal tract. These nutrients can be used by the microflora, resulting in the production of bacterial metabolites, for example, SCFA, which may affect intestinal gene expression indirectly. The present review provides an insight on possible effects of reduced feed intake on intestinal gene expression, as it may occur post-weaning. Detailed knowledge on effects of reduced feed intake on intestinal gene expression may help to understand weaning-associated intestinal dysfunctions and diseases. Examples are given of intestinal genes which may be altered in their expression due to supply with specific nutrients. In that way, gene expression could be modulated by dietary means, thereby acting as a potential therapeutic tool. This could be achieved, for example, by influencing genes coding for digestive or absorptive proteins, thus optimising digestive function and metabolism, but also with regard to immune response, or by influencing proliferative processes, thereby enhancing mucosal repair. This would be of special interest when designing a diet to overcome weaning-associated problems.
Collapse
|
30
|
Longevity in mice is promoted by probiotic-induced suppression of colonic senescence dependent on upregulation of gut bacterial polyamine production. PLoS One 2011; 6:e23652. [PMID: 21858192 PMCID: PMC3156754 DOI: 10.1371/journal.pone.0023652] [Citation(s) in RCA: 200] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 07/22/2011] [Indexed: 11/19/2022] Open
Abstract
Background Chronic low-grade inflammation is recognized as an important factor contributing to senescence and age-related diseases. In mammals, levels of polyamines (PAs) decrease during the ageing process; PAs are known to decrease systemic inflammation by inhibiting inflammatory cytokine synthesis in macrophages. Reductions in intestinal luminal PAs levels have been associated with intestinal barrier dysfunction. The probiotic strain Bifidobacterium animalis subsp. lactis LKM512 is known to increase intestinal luminal PA concentrations. Methodology/Principal Findings We supplemented the diet of 10-month-old Crj:CD-1 female mice with LKM512 for 11 months, while the controls received no supplementation. Survival rates were compared using Kaplan–Meier survival curves. LKM512-treated mice survived significantly longer than controls (P<0.001); moreover, skin ulcers and tumors were more common in the control mice. We then analyzed inflammatory and intestinal conditions by measuring several markers using HPLC, ELISA, reverse transcription-quantitative PCR, and histological slices. LKM512 mice showed altered 16S rRNA gene expression of several predominant intestinal bacterial groups. The fecal concentrations of PAs, but not of short-chain fatty acids, were significantly higher in LKM512-treated mice (P<0.05). Colonic mucosal function was also better in LKM512 mice, with increased mucus secretion and better maintenance of tight junctions. Changes in gene expression levels were evaluated using the NimbleGen mouse DNA microarray. LKM512 administration also downregulated the expression of ageing-associated and inflammation-associated genes and gene expression levels in 21-month-old LKM512-treated mice resembled those in 10-month-old untreated (younger) mice. Conclusion/Significance Our study demonstrated increased longevity in mice following probiotic treatment with LKM512, possibly due to the suppression of chronic low-grade inflammation in the colon induced by higher PA levels. This indicates that ingestion of specific probiotics may be an easy approach for improving intestinal health and increasing lifespan. Further studies are required to clarify its effectiveness in humans.
Collapse
|
31
|
Matsumoto M, Kurihara S. Probiotics-induced increase of large intestinal luminal polyamine concentration may promote longevity. Med Hypotheses 2011; 77:469-72. [PMID: 21745717 DOI: 10.1016/j.mehy.2011.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 06/06/2011] [Indexed: 11/27/2022]
Abstract
Many mechanisms contribute to senescence, such as telomere shortening in replicative cells, cumulative damage to DNA leading to genomic instability, and oxidative damage to molecules by reactive oxygen species (ROS). These include chronic low-grade inflammation (inflammageing), a major risk factor for ageing and age-related diseases, such as Alzheimer's disease and type II diabetes. Furthermore, the prevention of inflammageing seems to be one of the most effective approaches to increase longevity. Here, I discuss the rationale and recent evidence for probiotic-induced upregulation of intestinal luminal polyamine (PA) production in the extension of lifespan by preventing inflammageing.
Collapse
Affiliation(s)
- Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd., 20-1 Hirai, Hinode-cho, Nishitama-gun, Tokyo 190-0182, Japan.
| | | |
Collapse
|
32
|
Iwashita Y, Sakiyama T, Musch MW, Ropeleski MJ, Tsubouchi H, Chang EB. Polyamines mediate glutamine-dependent induction of the intestinal epithelial heat shock response. Am J Physiol Gastrointest Liver Physiol 2011; 301:G181-7. [PMID: 21512157 PMCID: PMC3129932 DOI: 10.1152/ajpgi.00054.2011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Heat shock proteins (Hsps) are highly conserved proteins that play a role in cytoprotection and maintaining intestinal homeostasis. Glutamine is essential for the optimal induction of intestinal epithelial Hsp expression, but its mechanisms of action are incompletely understood. Glutamine is a substrate for polyamine synthesis and stimulates the activity of ornithine decarboxylase (ODC), a key enzyme for polyamine synthesis, in intestinal epithelial cells. Thus we investigated whether polyamines (putrescine, spermidine, or spermine) and their precursor ornithine mediate the induction of Hsp expression in IEC-18 rat intestinal epithelial cells. As previously observed, glutamine was required for heat stress induction of Hsp70 and Hsp25, although it had little effect under basal conditions. Under conditions of glutamine depletion, supplementation of ornithine or polyamines restored the heat-induced expression of Hsp70 and Hsp25. When ODC was inhibited by α-difluoromethylornithine (DFMO), an irreversible ODC inhibitor, the heat stress induction of Hsp70 and Hsp25 was decreased significantly, even in the presence of glutamine. Ornithine, polyamines, and DFMO did not modify the nuclear localization of heat shock transcription factor 1 (HSF-1). However, DFMO dramatically reduced glutamine-dependent HSF-1 binding to an oligonucleotide with heat shock elements (HSE), which was increased by glutamine. In addition, exogenous polyamines recovered the DNA-binding activity. These results indicate that polyamines play a critical role in the glutamine-dependent induction of the intestinal epithelial heat shock response through facilitation of HSF-1 binding to HSE.
Collapse
Affiliation(s)
- Yuji Iwashita
- 1Department of Digestive and Life-style related Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Toshio Sakiyama
- 1Department of Digestive and Life-style related Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mark W. Musch
- 2Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Mark J. Ropeleski
- 3Gastrointestinal Diseases Research Unit, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Hirohito Tsubouchi
- 1Department of Digestive and Life-style related Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Eugene B. Chang
- 2Department of Medicine, University of Chicago, Chicago, Illinois; and
| |
Collapse
|
33
|
Abstract
Polyamines are organic cations shown to control gene expression at the transcriptional, posttranscriptional, and translational levels. Multiple cellular oncogenic pathways are involved in regulation of transcription and translation of polyamine-metabolizing enzymes. As a consequence of genetic alterations, expression levels and activities of polyamine-metabolizing enzymes change rapidly during tumorigenesis resulting in high levels of polyamines in many human epithelial tumors. This review summarizes the mechanisms of polyamine regulation by canonical tumor suppressor genes and oncogenes, as well as the role of eukaryotic initiation factor 5A (EIF5A) in cancer. The importance of research utilizing pharmaceutical inhibitors and cancer chemopreventive strategies targeting the polyamine pathway is also discussed.
Collapse
Affiliation(s)
- Edwin A Paz
- Cancer Biology Interdisciplinary Graduate Program, Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | | | | |
Collapse
|
34
|
Roskott AMC, Nieuwenhuijs VB, Dijkstra G, Koudstaal LG, Leuvenink HGD, Ploeg RJ. Small bowel preservation for intestinal transplantation: a review. Transpl Int 2010; 24:107-31. [PMID: 21083772 DOI: 10.1111/j.1432-2277.2010.01187.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intestinal transplantation has become the therapy of choice for patients with intestinal failure and life-threatening complications from total parenteral nutrition. Results, however, remain inferior as compared with other transplant types with the quality of the organ graft as the most important factor of outcome after transplantation. The intestine is extremely sensitive to ischemia. Unfortunately, a relatively long ischemic preservation period is inevitable. The current standard in organ preservation [cold storage (CS) with University of Wisconsin solution] was developed for kidney/liver preservation and is suboptimal for the intestinal graft despite good results for other organs. This review aimed at appraising the results from the use of previously applied and recently developed preservation solutions and techniques to identify key areas for improvement. As the studies available do not reveal the most effective method for intestinal preservation, an optimal strategy will result from a synergistic effect of different vital elements identified from a review of published material from the literature. A key factor is the composition of the solution using a low-viscosity solution to facilitate washout of blood, including amino acids to improve viability, impermeants and colloids to prevent edema, and buffer for pH-homeostasis. Optimizing conditions include a vascular flush before CS and luminal preservation. The most effective composition of the luminal solution and a practical, clinically applicable optimal technique are yet to reach finality. Short-duration oxygenated arterial and/or luminal perfusion have to be considered. Thus, a tailor-made approach to luminal preservation solution and technique need further investigation in transplant models and the human setting to develop the ultimate technique meeting the physiologic demands of the intestinal graft during preservation.
Collapse
|
35
|
Cho SO, Lim JW, Jun JH, Kim KH, Kim H. Helicobacter pylori in a Korean isolate expressed proteins differentially in human gastric epithelial cells. Dig Dis Sci 2010; 55:1550-64. [PMID: 19672714 DOI: 10.1007/s10620-009-0908-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Accepted: 07/05/2009] [Indexed: 12/19/2022]
Abstract
PURPOSE The proteins expressed in gastric epithelial cells infected with Helicobacter pylori (H. pylori) may determine the clinical outcome such as chronic gastritis, peptic ulcer, and gastric carcinoma. The present study aims to determine the differentially expressed proteins in human gastric epithelial AGS cells that were infected with H. pylori in a Korean isolate, a cagA+, vacA s1b m2 iceA1 H. pylori by proteomic analysis. The differentially expressed proteins, whose expression levels were more or less than twofold in H. pylori-infected cells, were analyzed. RESULTS Ten proteins (chromatin assembly factor-1, proliferating cell nuclear antigen, 14-3-3 protein tau, eukaryotic translation initiation factor 6, heat-shock protein 90beta, dimethylarginine dimethylaminohydrolase-1, L-lactate dehydrogenase B chain, prohibitin, triosephosphate isomerase, protein disulfide isomerase) were up-regulated while eight proteins (heat-shock gp96 precursor, nucleophosmin, ornithine aminotransferase, Ku70, L-arginine-glycine amidinotransferase, Smad anchor for receptor activation, ADP-ribosylation factor, WD repeat-containing protein isoform 1) were down-regulated by H. pylori infection in AGS cells. These proteins are related to cell proliferation, cell adhesion, carcinogenesis, cell-defense mechanisms against oxidative stress, membrane trafficking, and energy metabolism. CONCLUSIONS Oxidative stress, cell proliferation, cell adhesion, and membrane trafficking may be involved in the pathogenesis of gastric diseases including cancer associated with H. pylori in a Korean isolate.
Collapse
Affiliation(s)
- Soon Ok Cho
- Department of Pharmacology, Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
36
|
Xiao L, Rao JN, Zou T, Liu L, Yu TX, Zhu XY, Donahue JM, Wang JY. Induced ATF-2 represses CDK4 transcription through dimerization with JunD inhibiting intestinal epithelial cell growth after polyamine depletion. Am J Physiol Cell Physiol 2010; 298:C1226-34. [PMID: 20181929 DOI: 10.1152/ajpcell.00021.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intestinal epithelium is a rapidly self-renewing tissue in the body, and its homeostasis is tightly regulated by numerous factors including polyamines. Decreased levels of cellular polyamines increase activating transcription factor (ATF)-2, but the exact role and mechanism of induced ATF-2 in the regulation of intestinal epithelial cell (IEC) growth remain elusive. Cyclin-dependent kinase (CDK) 4 is necessary for the G1-to-S phase transition during the cell cycle, and its expression is predominantly controlled at the transcription level. Here, we reported that induced ATF-2 following polyamine depletion repressed CDK4 gene transcription in IECs by increasing formation of the ATF-2/JunD heterodimers. ATF-2 formed complexes with JunD as measured by immunoprecipitation using the ATF-2 and JunD antibodies and by glutathione S-transferase (GST) pull-down assays using GST-ATF-2 fusion proteins. Studies using various mutants of GST-ATF-2 revealed that formation of the ATF-2/JunD dimers depended on the COOH-terminal basic region-leucine zipper domain of ATF-2. Polyamine depletion increased ATF-2/JunD complex and inhibited CDK4 transcription as indicated by a decrease in the levels of CDK4-promoter activity and its mRNA. ATF-2 silencing not only prevented inhibition of CDK4 transcription in polyamine-deficient cells but also abolished repression of CDK4 expression induced by ectopic JunD overexpression. ATF-2 silencing also promoted IEC growth in polyamine-depleted cells. These results indicate that induced ATF-2/JunD association following polyamine depletion represses CDK4 transcription, thus contributing to the inhibition of IEC growth.
Collapse
Affiliation(s)
- Lan Xiao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Xie G, Zheng X, Qi X, Cao Y, Chi Y, Su M, Ni Y, Qiu Y, Liu Y, Li H, Zhao A, Jia W. Metabonomic evaluation of melamine-induced acute renal toxicity in rats. J Proteome Res 2010; 9:125-133. [PMID: 19476335 DOI: 10.1021/pr900333h] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The recent outbreak of renal failure in infants in China has been determined to be caused by melamine (Mel) and derivatives adulterated in the food. A metabonomic study was performed to evaluate the global biochemical alteration triggered by Mel ingestion in parallel with the acute renal toxicity in rats. Mel at 600, 300, and 100 mg/kg, cyanuric acid (Cya) at 100 mg/kg, and mixture of Mel and Cya (50 mg/kg each) were administered in five groups of Wistar rats, respectively, via oral gavage for 15 days. Urinary metabonomic profiles indicated that Mel perturbed urinary metabolism in a dose-dependent manner, with high-dose group showing the most significant impact. Metabonomic variations also suggest that the toxicity of low-dose (50 mg/kg) Mel was greatly elevated by the presence of Cya (at 50 mg/kg), which was able to induce a significant metabolic alteration to a level equivalent to that of 600 mg/kg Mel. Histological examination and serum biochemical analysis also indicated that the low-dose Mel-Cya mixture and high-dose Mel group resulted in the greatest renal toxicity. The high-dose Mel and low-dose Mel-Cya resulted in disrupted amino acid metabolism including tryptophan, polyamine, and tyrosine metabolism, and altered TCA and gut microflora structure.
Collapse
Affiliation(s)
- Guoxiang Xie
- Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Colorectal cancer chemoprevention, or chemoprophylaxis, is a drug-based approach to prevent colorectal cancer. Preventing colorectal adenomas with currently available agents demonstrates the promise of pharmacologic strategies directed at critical regulatory pathways. However, agent toxicity, lesion breakthrough and competing efficacy from endoscopy procedures challenge population-based implementation. This article reviews the role of colorectal cancer chemoprevention in the context of existing screening and surveillance guidelines and practice. Emphasis is placed on the role of the colorectal adenoma as a cancer precursor and its surrogacy in assessing individual risk and for evaluating chemoprevention efficacy. We discuss the importance of risk stratification for identifying subjects at moderate-to-high risk for colorectal cancer who are most likely to benefit from chemoprevention at an acceptable level of risk.
Collapse
Affiliation(s)
- Patricia A Thompson
- The University of Arizona, Gastrointestinal Cancer Program, Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, AZ 85724, USA
| | - Eugene W Gerner
- Cancer Prevention Pharmaceuticals, PO Box 36285, Tucson, AZ 85740, USA and The University of Arizona, Gastrointestinal Cancer Program, Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, AZ 85724, USA, Tel.: +1 520 626 2197, Fax: +1 520 626 4480,
| |
Collapse
|
39
|
Ishii K, Kono H, Hosomura N, Tsuchiya M, Ohgiku M, Tanaka N, Fujii H. Medium-chain triglycerides enhance mucous secretion and cell proliferation in the rat. J Gastroenterol 2009; 44:204-11. [PMID: 19214664 DOI: 10.1007/s00535-008-2308-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Accepted: 09/29/2008] [Indexed: 02/04/2023]
Abstract
BACKGROUND The specific purpose of this study was to investigate the effects of medium-chain triglycerides (MCTs) on intestinal cell proliferation and mucous secretion of the small intestine in the rat. METHODS Rats were fed chow diet and given MCTs or the same weight of corn oil (5 g/kg per day) by gavage daily for 2 weeks, and then tissue samples of the small intestines were harvested. Leptin concentration in the small intestine was measured. Cell proliferation and apoptosis in the small intestine was determined by immunohistochemistry. Diamine oxidase (DAO) activity was measured by colorimetric assay. RESULTS In rats fed only chow diet (normal rats), the number of goblet cells per villi was 14.2 +/- 0.75 in the jejunum and 15.2 +/- 1.12 in the ileum. The number of goblet cells increased significantly in rats given MCTs compared with rats given corn oil or normal rats. Ki-67-positive cells were detected on the entire villi and the crypts in the small intestine. Furthermore, the proliferative index and the apoptotic index were also significantly greater in rats given MCTs than rats given corn oil or normal rats. Moreover, DAO activity and leptin concentration in the small intestine were significantly greater in rats given MCTs than rats given corn oil or normal rats. CONCLUSIONS MCTs enhance cell proliferation of the intestinal epithelium and mucous secretion from goblet cells in the small intestine. These effects may protect the gut in patients suffering from inflammatory bowel disease or enterogenous infection.
Collapse
Affiliation(s)
- Kenichi Ishii
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Tjabringa GS, Zandieh-Doulabi B, Helder MN, Knippenberg M, Wuisman PIJM, Klein-Nulend J. The polymine spermine regulates osteogenic differentiation in adipose stem cells. J Cell Mol Med 2008; 12:1710-7. [PMID: 18194460 PMCID: PMC3918087 DOI: 10.1111/j.1582-4934.2008.00224.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
For bone tissue engineering, it is important that mesenchymal stem cells (MSCs) differentiate into osteoblasts. To develop a method for differentiation of adipose tissue-derived mesenchymal stem cells (AT-MSCs) along the osteogenic lineage, we studied the effect of polyamines, which are organic cations implicated in bone growth and development, on differentiation of AT-MSCs. Treatment of goat-derived AT-MSCs with 1,25-dihydroxyvitamin-D3 (1,25(OH)2D3), which stimulates osteogenic differentiation, for 7 days induced gene expression of the polyamine-modulated transcription factor-1 (PMF-1) and spermidine/spermine N (1)-acetyltransferase (SSAT), which are both involved in polyamine metabolism, suggesting that polyamines are involved in osteogenic differentiation of AT-MSCs. Furthermore, treatment of AT-MSCs with the polyamine spermine-regulated gene expression of runx-2, a transcription factor involved in early stages of osteogenic differentiation, and that of osteopontin, a bone matrix protein expressed in later stages of osteogenic differentiation. Runx-2 gene expression was increased 4 and 14 days after a short 30 min. treatment with spermine, while osteopontin gene expression was only increased 4 days after spermine treatment. Finally, alkaline phosphatase activity, which is intimately involved in the formation of extracellular matrix of bone, was increased 4 weeks after the 30 min.-spermine treatment of AT-MSCs. In conclusion, this study shows for the first time that the polyamine spermine regulates differentiation of AT-MSCs along the osteogenic lineage, which can be used as a new method for differentiation of AT-MSCs along the osteogenic lineage. Therefore, polyamines may constitute a promising tool for bone tissue engineering approaches using AT-MSCs, such as a one-step surgical procedure for spinal interbody fusion.
Collapse
Affiliation(s)
- G S Tjabringa
- Department of Oral Cell Biology, Academic Center of Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit, Research Institute MOVE, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
41
|
Yellepeddi VK, Pisal DS, Kumar A, Kaushik RS, Hildreth MB, Guan X, Palakurthi S. Permeability of surface-modified polyamidoamine (PAMAM) dendrimers across Caco-2 cell monolayers. Int J Pharm 2007; 350:113-21. [PMID: 17913410 PMCID: PMC2266586 DOI: 10.1016/j.ijpharm.2007.08.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 08/17/2007] [Accepted: 08/21/2007] [Indexed: 11/25/2022]
Abstract
Aim of this study was to prepare polyamine-conjugated PAMAM dendrimers and study their permeability across Caco-2 cell monolayers. Polyamines, namely, arginine and ornithine were conjugated to the amine terminals of the G4 PAMAM dendrimers by Fmoc synthesis. The apical-to-basolateral (AB) and basolateral-to-apical (BA) apparent permeability coefficients (P(app)) for the PAMAM dendrimers increased by conjugating the dendrimers with both of the polyamines. The enhancement in permeability was dependent on the dendrimer concentration and duration of incubation. The correlation between monolayer permeability and the decrease in transepithelial electrical resistance (TEER) with both the PAMAM dendrimers and the polyamine-conjugated dendrimers suggests that paracellular transport is one of the mechanisms of transport across the epithelial cells. Cytotoxicity of the polyamine-conjugated dendrimers was evaluated in Caco-2 cells by MTT (methylthiazoletetrazolium) assay. Arginine-conjugated dendrimers were slightly more toxic than PAMAM dendrimer as well as ornithine-conjugated dendrimers. Though investigations on the possible involvement of other transport mechanisms are in progress, results of the present study suggest the potential of dendrimer-polyamine conjugates as drug carriers to increase the oral absorption of drugs.
Collapse
Affiliation(s)
- Venkata K. Yellepeddi
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Dipak S. Pisal
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Ajay Kumar
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Radhey S. Kaushik
- Department of Biology & Microbiology/Veterinary Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Michael B. Hildreth
- Department of Biology & Microbiology, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Xiangming Guan
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, 1 Administration Lane, South Dakota State University, Brookings, SD 57007
| |
Collapse
|
42
|
Wild GE, Searles LE, Koski KG, Drozdowski LA, Begum-Hasan J, Thomson ABR. Oral polyamine administration modifies the ontogeny of hexose transporter gene expression in the postnatal rat intestine. Am J Physiol Gastrointest Liver Physiol 2007; 293:G453-60. [PMID: 17673438 DOI: 10.1152/ajpgi.00077.2006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastrointestinal mucosal polyamines influence enterocyte proliferation and differentiation during small intestinal maturation in the rat. Studies in postnatal rats have shown that ornithine decarboxylase (ODC) protein and mRNA peak before the maximal expression of brush-border membrane (BBM) sucrase-isomaltase (SI) and the sugar transporters sodium-dependent glucose transporter 1 (SGLT1) and glucose transporter 2 (GLUT2). This study was undertaken to test the hypothesis that the oral administration of spermidine in postnatal rats upregulates the expression of ODC, thereby enhancing the expression of SI and SGLT1 in the brush-border membrane as well as basolateral membrane-facilitative GLUT2 and Na(+)-K(+)-ATPase. Northern and Western blot analyses were performed with antibodies and cDNA probes specific for SI, SGLT1, GLUT2, alpha(1)- and beta(1)-subunits of Na(+)-K(+)-ATPase, and ODC. Postnatal rats fed 6 mumol spermidine daily for 3 days from days 7 to 9 were killed either on postnatal day 10 (Sp10) or day 13 following a 3-day washout period (Sp13). Sp10 rats showed a precocious increase in the abundance of mRNAs for SI, SGLT1, and GLUT2 and Na(+)-K(+)-ATPase activity and alpha(1)- and beta(1)-isoform gene expression compared with controls. ODC activity and protein and mRNA abundance were also increased in Sp10 animals. The increased expression of these genes was not sustained in Sp13 rats, suggesting that these effects were transient. Thus, 3 days of oral polyamine administration induces the precocious maturation of glucose transporters in the postnatal rat small intestine, which may be mediated by alterations in ODC expression.
Collapse
Affiliation(s)
- G E Wild
- Division of Gastroenterology, Department of Medicine, McGill University Health Center, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Ulrich S, Huwiler A, Loitsch S, Schmidt H, Stein JM. De novo ceramide biosynthesis is associated with resveratrol-induced inhibition of ornithine decarboxylase activity. Biochem Pharmacol 2007; 74:281-289. [PMID: 17521618 DOI: 10.1016/j.bcp.2007.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 03/30/2007] [Accepted: 04/02/2007] [Indexed: 11/25/2022]
Abstract
Previous studies could demonstrate, that the naturally occuring polyphenol resveratrol inhibits cell growth of colon carcinoma cells at least in part by inhibition of protooncogene ornithine decarboxylase (ODC). The objective of this study was to provide several lines of evidence suggesting that the induction of ceramide synthesis is involved in this regulatory mechanisms. Cell growth was determined by BrdU incorporation and crystal violet staining. Ceramide concentrations were detected by HPLC-coupled mass-spectrometry. Protein levels were examined by Western blot analysis. ODC activity was assayed radiometrically measuring [(14)CO(2)]-liberation. A dominant-negative PPARgamma mutant was transfected in Caco-2 cells to suppress PPARgamma-mediated functions. Antiproliferative effects of resveratrol closely correlate with a dose-dependent increase of endogenous ceramides (p<0.001). Compared to controls the cell-permeable ceramide analogues C2- and C6-ceramide significantly inhibit ODC-activity (p<0.001) in colorectal cancer cells. C6-ceramide further diminished protein levels of protooncogenes c-myc (p<0.05) and ODC (p<0.01), which is strictly related to the ability of ceramides to inhibit cell growth in a time- and dose-dependent manner. These results were further confirmed using inhibitors of sphingolipid metabolism, where only co-incubation with a serine palmitoyltransferase (SPT) inhibitor could significantly counteract resveratrol-mediated actions. These data suggest that the induction of ceramide de novo biosynthesis but not hydrolysis of sphingomyelin is involved in resveratrol-mediated inhibition of ODC. In contrast to the regulation of catabolic spermidine/spermine acetyltransferase by resveratrol, inhibitory effects on ODC occur PPARgamma-independently, indicating independent pathways of resveratrol-action. Due to our findings resveratrol could show great chemopreventive and therapeutic potential in the treatment of colorectal cancers.
Collapse
Affiliation(s)
- Sandra Ulrich
- First Department of Internal Medicine-ZAFES, Johann Wolfgang Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
44
|
Fischer da Silva AV, Borges SA, Maiorka A, Givisiez PEN, Rocha C, Macari M. Ornithine decarboxylase expression in the small intestine of broilers submitted to feed restriction and glutamine supplementation. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2007. [DOI: 10.1590/s1516-635x2007000200006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
45
|
Xiao L, Rao J, Zou T, Liu L, Marasa B, Chen J, Turner D, Passaniti A, Wang JY. Induced JunD in intestinal epithelial cells represses CDK4 transcription through its proximal promoter region following polyamine depletion. Biochem J 2007; 403:573-81. [PMID: 17253961 PMCID: PMC1876376 DOI: 10.1042/bj20061436] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 01/17/2007] [Accepted: 01/25/2007] [Indexed: 02/06/2023]
Abstract
Maintenance of intestinal epithelial integrity requires cellular polyamines that regulate expression of various genes involved in cell proliferation, growth arrest and apoptosis. In prior studies, depletion of cellular polyamines has been shown to stabilize JunD, a member of the AP-1 (activator protein-1) family of transcription factors, leading to inhibition of intestinal epithelial cell proliferation, but the exact downstream targets of induced JunD remain elusive. CDK4 (cyclin-dependent kinase 4) is essential for the G1- to S-phase transition during the cell cycle and its expression is primarily controlled at the transcriptional level. In the present study, we show that induced JunD in IECs (intestinal epithelial cells) is a transcriptional repressor of the CDK4 gene following polyamine depletion. Increased JunD in polyamine-deficient cells was associated with a significant inhibition of CDK4 transcription, as indicated by repression of CDK4-promoter activity and decreased levels of CDK4 mRNA and protein, all of which were prevented by using specific antisense JunD oligomers. Ectopic expression of the wild-type junD also repressed CDK4-promoter activity and decreased levels of CDK4 mRNA and protein without any effect on CDK2 expression. Gel shift and chromatin immunoprecipitation assays revealed that JunD bound to the proximal region of the CDK4-promoter in vitro as well as in vivo, while experiments using different CDK4-promoter mutants showed that transcriptional repression of CDK4 by JunD was mediated through an AP-1 binding site within this proximal sequence of the CDK4-promoter. These results indicate that induced JunD in IECs represses CDK4 transcription through its proximal promoter region following polyamine depletion.
Collapse
Key Words
- activator protein-1 (ap-1)
- α-difluoromethylornithine
- growth arrest
- intestinal epithelium
- ornithine decarboxylase
- transcriptional regulation
- ap-1, activator protein-1
- cdk, cyclin-dependent kinase
- chip, chromatin immunoprecipitation
- dfmo, α-difluoromethylornithine
- emsa, electrophoretic mobility-shift assay
- fbs, fetal bovine serum
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- iec, intestinal epithelial cell
- luc, luciferase
- pbs-t, pbs containing tween 20
- q-pcr, quantitative pcr
- rb, retinoblastoma tumour suppressor protein
- rt, reverse transcriptase
Collapse
Affiliation(s)
- Lan Xiao
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jaladanki N. Rao
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Tongtong Zou
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Lan Liu
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Bernard S. Marasa
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- †Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jie Chen
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Douglas J. Turner
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Antonino Passaniti
- †Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| | - Jian-Ying Wang
- *Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- †Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, U.S.A
- ‡Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, U.S.A
| |
Collapse
|
46
|
Lenaerts K, Bouwman FG, Lamers WH, Renes J, Mariman EC. Comparative proteomic analysis of cell lines and scrapings of the human intestinal epithelium. BMC Genomics 2007; 8:91. [PMID: 17407598 PMCID: PMC1852558 DOI: 10.1186/1471-2164-8-91] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Accepted: 04/03/2007] [Indexed: 12/31/2022] Open
Abstract
Background In vitro models are indispensable study objects in the fields of cell and molecular biology, with advantages such as accessibility, homogeneity of the cell population, reproducibility, and growth rate. The Caco-2 cell line, originating from a colon carcinoma, is a widely used in vitro model for small intestinal epithelium. Cancer cells have an altered metabolism, making it difficult to infer their representativity for the tissue from which they are derived. This study was designed to compare the protein expression pattern of Caco-2 cells with the patterns of intestinal epithelial cells from human small and large intestine. HT-29 intestinal cells, Hep G2 liver cells and TE 671 muscle cells were included too, the latter two as negative controls. Results Two-dimensional gel electrophoresis was performed on each tissue and cell line protein sample. Principal component and cluster analysis revealed that global expression of intestinal epithelial scrapings differed from that of intestinal epithelial cell lines. Since all cultured cell lines clustered together, this finding was ascribed to an adaptation of cells to culture conditions and their tumor origin, and responsible proteins were identified by mass spectrometry. When investigating the profiles of Caco-2 cells and small intestinal cells in detail, a considerable overlap was observed. Conclusion Numerous proteins showed a similar expression in Caco-2 cells, HT-29 cells, and both the intestinal scrapings, of which some appear to be characteristic to human intestinal epithelium in vivo. In addition, several biologically significant proteins are expressed at comparable levels in Caco-2 cells and small intestinal scrapings, indicating the usability of this in vitro model. Caco-2 cells, however, appear to over-express as well as under-express certain proteins, which needs to be considered by scientists using this cell line. Hence, care should be taken to prevent misinterpretation of in vitro obtained findings when translating them to the in vivo situation.
Collapse
Affiliation(s)
- Kaatje Lenaerts
- Maastricht Proteomics Center, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Department of Human Biology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Freek G Bouwman
- Maastricht Proteomics Center, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Department of Human Biology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Wouter H Lamers
- AMC Liver Center, Academic Medical Center, University of Amsterdam, Meibergdreef 69, 1105 BK Amsterdam, The Netherlands
| | - Johan Renes
- Maastricht Proteomics Center, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Department of Human Biology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Edwin C Mariman
- Maastricht Proteomics Center, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Department of Human Biology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
47
|
Romero MJ, Platt DH, Caldwell RB, Caldwell RW. Therapeutic use of citrulline in cardiovascular disease. ACTA ACUST UNITED AC 2007; 24:275-90. [PMID: 17214603 DOI: 10.1111/j.1527-3466.2006.00275.x] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
L-citrulline is the natural precursor of L-arginine, substrate for nitric oxide synthase (NOS) in the production of NO. Supplemental administration L-arginine has been shown to be effective in improving NO production and cardiovascular function in cardiovascular diseases associated with endothelial dysfunction, such as hypertension, heart failure, atherosclerosis, diabetic vascular disease and ischemia-reperfusion injury, but the beneficial actions do not endure with chronic therapy. Substantial intestinal and hepatic metabolism of L-arginine to ornithine and urea by arginase makes oral delivery very ineffective. Additionally, all of these disease states as well as supplemental L-arginine enhance arginase expression and activity, thus reducing the effectiveness of L-arginine therapy. In contrast, L-citrulline is not metabolized in the intestine or liver and does not induce tissue arginase, but rather inhibits its activity. L-citrulline entering the kidney, vascular endothelium and other tissues can be readily converted to L-arginine, thus raising plasma and tissue levels of L-arginine and enhancing NO production. Supplemental L-citrulline has promise as a therapeutic adjunct in disease states associated with L-arginine deficiencies.
Collapse
Affiliation(s)
- Maritza J Romero
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta GA, USA
| | | | | | | |
Collapse
|
48
|
Rao JN, Liu L, Zou T, Marasa BS, Boneva D, Wang SR, Malone DL, Turner DJ, Wang JY. Polyamines are required for phospholipase C-gamma1 expression promoting intestinal epithelial restitution after wounding. Am J Physiol Gastrointest Liver Physiol 2007; 292:G335-43. [PMID: 16973916 DOI: 10.1152/ajpgi.00282.2006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal mucosal restitution occurs by epithelial cell migration, rather than by proliferation, to reseal superficial wounds after injury. Polyamines are essential for the stimulation of intestinal epithelial cell (IEC) migration during restitution in association with their ability to regulate Ca2+ homeostasis, but the exact mechanism by which polyamines induce cytosolic free Ca2+ concentration ([Ca2+]cyt) remains unclear. Phospholipase C (PLC)-gamma1 catalyzes the formation of inositol (1,4,5)-trisphosphate (IP3), which is implicated in the regulation of [Ca2+]cyt by modulating Ca2+ store mobilization and Ca2+ influx. The present study tested the hypothesis that polyamines are involved in PLC-gamma1 activity, regulating [Ca2+]cyt and cell migration after wounding. Depletion of cellular polyamines by alpha-difluoromethylornithine inhibited PLC-gamma1 expression in differentiated IECs (stable Cdx2-transfected IEC-6 cells), as indicated by substantial decreases in levels of PLC-gamma1 mRNA and protein and its enzyme product IP3. Polyamine-deficient cells also displayed decreased [Ca2+]cyt and inhibited cell migration. Decreased levels of PLC-gamma1 by treatment with U-73122 or transfection with short interfering RNA specifically targeting PLC-gamma1 also decreased IP3, reduced resting [Ca2+]cyt and Ca2+ influx after store depletion, and suppressed cell migration in control cells. In contrast, stimulation of PLC-gamma1 by 2,4,6-trimethyl-N-(meta-3-trifluoromethylphenyl)-benzenesulfonamide induced IP3, increased [Ca2+]cyt, and promoted cell migration in polyamine-deficient cells. These results indicate that polyamines are absolutely required for PLC-gamma1 expression in IECs and that polyamine-mediated PLC-gamma1 signaling stimulates cell migration during restitution as a result of increased [Ca2+]cyt.
Collapse
Affiliation(s)
- Jaladanki N Rao
- Cell Biology Group, Department of Surgery, and 2Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tjabringa GS, Vezeridis PS, Zandieh-Doulabi B, Helder MN, Wuisman PIJM, Klein-Nulend J. Polyamines Modulate Nitric Oxide Production andCox-2Gene Expression in Response to Mechanical Loading in Human Adipose Tissue-Derived Mesenchymal Stem Cells. Stem Cells 2006; 24:2262-9. [PMID: 16794268 DOI: 10.1634/stemcells.2005-0625] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
For bone tissue engineering, it is important that mesenchymal stem cells (MSCs) display a bone cell-like response to mechanical loading. We have shown earlier that this response includes increased nitric oxide (NO) production and cyclooxygenase-2 (COX-2) gene expression, both of which are intimately involved in mechanical adaptation of bone. COX-2 gene expression is likely regulated by polyamines, which are organic cations implicated in cell proliferation and differentiation. This has led to the hypothesis that polyamines may play a role in the response of adipose tissue-derived MSCs (AT-MSCs) to mechanical loading. The aim of this study was to investigate whether genes involved in polyamine metabolism are regulated by mechanical loading and to study whether polyamines modulate mechanical loading-induced NO production and COX-2 gene expression in human AT-MSCs. Human AT-MSCs displayed a bone cell-like response to mechanical loading applied by pulsating fluid flow (PFF), as demonstrated by increased NO production and increased gene expression of COX-2. Furthermore, PFF increased gene expression of spermidine/spermine N (1)-acetyltransferase, which is involved in polyamine catabolism, suggesting that mechanical loading modulates polyamine levels. Finally, the polyamine spermine was shown to inhibit both PFF-induced NO production and COX-2 gene expression, suggesting that polyamines modulate the response of human AT-MSCs to mechanical loading. In conclusion, this is the first study implicating polyamines in the response of human AT-MSCs to mechanical loading, creating opportunities for the use of polyamines in tissue engineering approaches targeting skeletal defects.
Collapse
Affiliation(s)
- Geuranne S Tjabringa
- Department of Oral Cell Biology, Academic Center of Dentistry Amsterdam, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
50
|
Gonzalez-Esquerra R, Leeson S. Concentrations of putrescine, spermidine, and spermine in duodenum and pancreas as affected by the ratio of arginine to lysine and source of methionine in broilers under heat stress. Poult Sci 2006; 85:1398-408. [PMID: 16903470 DOI: 10.1093/ps/85.8.1398] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
An experiment was designed to investigate the effect of Arg, Lys, Met, and environmental temperature on broiler performance and associated changes in duodenal and pancreatic polyamines. Two groups of 26-d-old Ross male broilers raised under thermoneutral (TN) conditions were reallocated to 4 rooms kept at heat stress (HS) or TN. Birds were fed equimolar amounts of 2-hydroxy-4-(methylthio) butanoic acid (HMB) or DL-Met (DLM) at requirement levels with Arg:Lys at 0.95 or 1.40. Twelve replicates of 4 birds were offered each diet ad libitum. Body weight gain, efficiency of dietary CP accretion (CPE), feed intake, and feed conversion ratio were ascertained from 26 to 33 d and from 34 to 47 d of age. One bird per cage was killed at 33 and 47 d, and samples of duodenum and pancreas were assayed for putrescine, spermidine, and spermine (Spm), together with estimates of duodenal villus height. From 26 to 33 d, birds fed HMB performed better than those fed DLM, but only at TN conditions. From 34 to 47 d, feeding HMB tended to optimize CPE when added to diets high in Arg. However, lower CPE was obtained when HMB was added to low-Arg diets, whereas birds fed DLM were unaffected by these treatments (P < 0.10). Methionine source, Arg:Lys, or both affected the concentrations of duodenal and pancreatic polyamines, with some changes correlating with performance variables during HS (P > 0.05). It was found that HS caused lower tissue spermidine (P < 0.001) and higher pancreatic Spm (P = 0.08) from 34 to 47 d. Putrescine concentrations were affected by diet and HS, depending on tissue and experimental period. Pancreatic Spm correlated negatively with changes in CPE influenced by Arg:Lys by Met source interaction in chronically heat-stressed birds. The possible association between polyamine metabolism and some of the effects of the Arg:Lys by Met source interaction observed in chronically stressed birds deserves further investigation.
Collapse
|