1
|
Wathanavasin W, Cheungpasitporn W, Thongprayoon C, Fülöp T. Effects of Dietary Fiber Supplementation on Modulating Uremic Toxins and Inflammation in Chronic Kidney Disease Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Toxins (Basel) 2025; 17:57. [PMID: 39998074 PMCID: PMC11860371 DOI: 10.3390/toxins17020057] [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: 01/01/2025] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Emerging evidence supports the beneficial effects of dietary fiber supplementation in alleviating gut dysbiosis, which leads to a reduction in uremic toxins and inflammatory markers in chronic kidney disease (CKD) patients. However, current evidence-based renal nutrition guidelines do not provide recommendations regarding dietary fiber intake. We performed a systematic review and meta-analysis to investigate and highlight the effects of dietary fiber supplementation on modulating uremic toxins and inflammatory markers in individuals with CKD, with or without dialysis. The eligible randomized controlled trials (RCTs) were identified from PubMed, Scopus, and Cochrane Central Register of Controlled trials until 27 November 2024. The results were synthesized using a random-effects model and presented as standardized mean differences (SMDs) with a 95% confidence interval (CI). A total of 21 studies with 700 patients were included. When compared with the control group, dietary fiber supplementation ranging from 6 to 50 g/day, for typically more than 4 weeks, could significantly reduce the levels of serum uremic toxins, including p-cresyl sulfate, indoxyl sulfate, and blood urea nitrogen (SMD -0.22, -0.34, -0.25, respectively, with p-values < 0.05), as well as biomarkers of inflammation, including interleukin-6 and tumor necrosis factor alpha (SMD -0.44, -0.34, respectively, with p-values < 0.05). These beneficial effects were consistent across different types of fibers and CKD status (with or without dialysis). However, no significant reduction in serum trimethylamine N-oxide, uric acid, and high-sensitivity C-reactive protein levels was observed with dietary fiber intervention. This study would pave the way for prioritizing dietary quality, particularly a fiber-rich diet, beyond the traditional focus on the quantities of protein, energy, and electrolyte restrictions among individuals with CKD.
Collapse
Affiliation(s)
- Wannasit Wathanavasin
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (W.W.); (W.C.); (C.T.)
- Nephrology Unit, Department of Medicine, Charoenkrung Pracharak Hospital, Bangkok Metropolitan Administration, Bangkok 10120, Thailand
| | - Wisit Cheungpasitporn
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (W.W.); (W.C.); (C.T.)
| | - Charat Thongprayoon
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (W.W.); (W.C.); (C.T.)
| | - Tibor Fülöp
- Medicine Service, Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Zhou M, Wu J, Wu L, Sun X, Chen C, Huang L. The utilization of N-acetylgalactosamine and its effect on the metabolism of amino acids in Erysipelotrichaceae strain. BMC Microbiol 2024; 24:397. [PMID: 39379811 PMCID: PMC11462708 DOI: 10.1186/s12866-024-03505-z] [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: 05/01/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The metabolism of gut microbiota produces bioactive metabolites that modulate host physiology and promote self-growth. Erysipelotrichaceae is one of the most common anaerobic microorganism families in the gut, which has been discovered to play a vital role in host metabolic disorders and inflammatory diseases. Our previous study found that N-acetylgalactosamine (GalNAc) in caecal content of pigs significantly affected the abundance of Erysipelotrichaceae strains. However, it remains unknown how GalNAc feeding in vitro culture affects the expression levels of genes in the GalNAc metabolic pathway and the concentrations of intermediate metabolites in the Erysipelotrichaceae strain. Whether GalNAc feeding should influence the metabolism of other nutrients, such as amino acids, remains unrevealed. RESULTS In this study, whole-genome sequence, transcriptome, and metabolome data were analyzed to assess the utilization of a Erysipelotrichaceae strain on GalNAc. The results showed the presence of a complete GalNAc catabolism pathway in the genome of this Erysipelotrichaceae strain. GalNAc feeding to this Erysipelotrichaceae strain significantly changed the expression levels of genes involved in glycolysis and tricarboxylic acid (TCA) cycle. Meanwhile, the concentrations of lactate, pyruvate, citrate, succinate and malate from the glycolysis and TCA cycle were significantly increased. In addition, transcriptome analysis indicated that the genes involved in the metabolism of amino acids were affected by GalNAc, including lysA (a gene involved in lysine biosynthesis) that was significantly down-regulated. The intracellular concentrations of 14 amino acids in the Erysipelotrichaceae strain were significantly increased after feeding GalNAc. CONCLUSIONS Our findings comfirmed and extended our previous works that demonstrated the utilization of GalNAc by Erysipelotrichaceae strain, and explained the possible mechanism of GalNAc affecting the abundance of Erysipelotrichaceae strain in vitro.
Collapse
Affiliation(s)
- Mengqing Zhou
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi Province, PR China
| | - Jinyuan Wu
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi Province, PR China
| | - Lin Wu
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi Province, PR China
| | - Xiao Sun
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi Province, PR China
| | - Congying Chen
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi Province, PR China.
| | - Lusheng Huang
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi Province, PR China.
| |
Collapse
|
3
|
Jiang L, Han D, Hao Y, Song Z, Sun Z, Dai Z. Linking serotonin homeostasis to gut function: Nutrition, gut microbiota and beyond. Crit Rev Food Sci Nutr 2024; 64:7291-7310. [PMID: 36861222 DOI: 10.1080/10408398.2023.2183935] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Serotonin (5-HT) produced by enterochromaffin (EC) cells in the digestive tract is crucial for maintaining gut function and homeostasis. Nutritional and non-nutritional stimuli in the gut lumen can modulate the ability of EC cells to produce 5-HT in a temporal- and spatial-specific manner that toning gut physiology and immune response. Of particular interest, the interactions between dietary factors and the gut microbiota exert distinct impacts on gut 5-HT homeostasis and signaling in metabolism and the gut immune response. However, the underlying mechanisms need to be unraveled. This review aims to summarize and discuss the importance of gut 5-HT homeostasis and its regulation in maintaining gut metabolism and immune function in health and disease with special emphasis on different types of nutrients, dietary supplements, processing, and gut microbiota. Cutting-edge discoveries in this area will provide the basis for the development of new nutritional and pharmaceutical strategies for the prevention and treatment of serotonin homeostasis-related gut and systematic disorders and diseases.
Collapse
Affiliation(s)
- Lili Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Youling Hao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhuan Song
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhiyuan Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
4
|
Liu Y, Zhang J, Yang G, Tang C, Li X, Lu L, Long K, Sun J, Ding Y, Li X, Li M, Ge L, Ma J. Effects of the commensal microbiota on spleen and mesenteric lymph node immune function: investigation in a germ-free piglet model. Front Microbiol 2024; 15:1398631. [PMID: 38933022 PMCID: PMC11201156 DOI: 10.3389/fmicb.2024.1398631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Commensal microbial-host interaction is crucial for host metabolism, growth, development, and immunity. However, research on microbial-host immunity in large animal models has been limited. This study was conducted to investigate the effects of the commensal microbiota on immune function in two model groups: germ-free (GF) and specific-pathogen-free (SPF) piglets. The weight and organ index of the spleen of the GF piglet were larger than those in the SPF piglet (P < 0.05). The histological structure of the red pulp area and mean area of germinal centers were larger in the SPF piglet than in the GF piglet (P < 0.05), whereas the areas of staining of B cells and T cells in the spleen and mesenteric lymph nodes (MLNs) were lower in the GF piglet (P < 0.05). We identified immune-related genes in the spleen and MLNs using RNA sequencing, and used real-time quantitative PCR to analyze the expression of core genes identified in gene set enrichment analysis. The expression levels of genes in the transforming growth factor-β/SMAD3 signaling pathway, Toll-like receptor 2/MyD88/nuclear factor-κB signaling pathway, and pro-inflammatory factor genes IL-6 and TNF-α in the spleen and MLNs were higher in the SPF piglet and in splenic lymphocytes compared with those in the GF and control group, respectively, under treatment with acetic acid, propionic acid, butyric acid, lipopolysaccharide (LPS), or concanavalin A (ConA). The abundances of plasma cells, CD8++ T cells, follicular helper T cells, and resting natural killer cells in the spleen and MLNs were significantly greater in the SPF piglet than in the GF piglet (P < 0.05). In conclusion, the commensal microbiota influenced the immune tissue structure, abundances of immune cells, and expression of immune-related pathways, indicating the importance of the commensal microbiota for spleen and MLNs development and function. In our study, GF piglet was used as the research model, eliminating the interference of microbiota in the experiment, and providing a suitable and efficient large animal research model for exploring the mechanism of "microbial-host" interactions.
Collapse
Affiliation(s)
- Yan Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Guitao Yang
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Chuang Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiaokai Li
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Lu Lu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Keren Long
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Yuchun Ding
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, China
| | - Jideng Ma
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Chongqing Academy of Animal Sciences, Chongqing, China
| |
Collapse
|
5
|
Arturo Tozzi, Minella R. Dynamics and metabolic effects of intestinal gases in healthy humans. Biochimie 2024; 221:81-90. [PMID: 38325747 DOI: 10.1016/j.biochi.2024.02.001] [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: 11/04/2023] [Revised: 01/06/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
Many living beings use exogenous and/or endogenous gases to attain evolutionary benefits. We make a comprehensive assessment of one of the major gaseous reservoirs in the human body, i.e., the bowel, providing extensive data that may serve as reference for future studies. We assess the intestinal gases in healthy humans, including their volume, composition, source and local distribution in proximal as well as distal gut. We analyse each one of the most abundant intestinal gases including nitrogen, oxygen, nitric oxide, carbon dioxide, methane, hydrogen, hydrogen sulfide, sulfur dioxide and cyanide. For every gas, we describe diffusive patterns, active trans-barrier transport dynamics, chemical properties, intra-/extra-intestinal metabolic effects mediated by intracellular, extracellular, paracrine and distant actions. Further, we highlight the local and systemic roles of gasotransmitters, i.e., signalling gaseous molecules that can freely diffuse through the intestinal cellular membranes. Yet, we provide testable hypotheses concerning the still unknown effects of some intestinal gases on the myenteric and submucosal neurons.
Collapse
Affiliation(s)
- Arturo Tozzi
- Center for Nonlinear Science, Department of Physics, University of North Texas, 1155 Union Circle, #311427, Denton, TX, 76203-5017, USA.
| | | |
Collapse
|
6
|
Taylor-Bowden T, Bhogoju S, Khwatenge CN, Nahashon SN. The Impact of Essential Amino Acids on the Gut Microbiota of Broiler Chickens. Microorganisms 2024; 12:693. [PMID: 38674637 PMCID: PMC11052162 DOI: 10.3390/microorganisms12040693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
The research involving the beneficial aspects of amino acids being added to poultry feed pertaining to performance, growth, feed intake, and feed conversion ratio is extensive. Yet currently the effects of amino acids on the gut microbiota aren't fully understood nor have there been many studies executed in poultry to explain the relationship between amino acids and the gut microbiota. The overall outcome of health has been linked to bird gut health due to the functionality of gastrointestinal tract (GIT) for digestion/absorption of nutrients as well as immune response. These essential functions of the GI are greatly driven by the resident microbiota which produce metabolites such as butyrate, propionate, and acetate, providing the microbiota a suitable and thrive driven environment. Feed, age, the use of feed additives and pathogenic infections are the main factors that have an effect on the microbial community within the GIT. Changes in these factors may have potential effects on the gut microbiota in the chicken intestine which in turn may have an influence on health essentially affecting growth, feed intake, and feed conversion ratio. This review will highlight limited research studies that investigated the possible role of amino acids in the gut microbiota composition of poultry.
Collapse
Affiliation(s)
- Thyneice Taylor-Bowden
- Department of Agriculture and Environmental Sciences, Tennessee State University, Nashville, TN 37209, USA;
| | - Sarayu Bhogoju
- College of Medicine, University of Kentucky, Lexington, KY 40506, USA;
| | - Collins N. Khwatenge
- College of Agriculture, Science and Technology, Department of Biological Sciences, Delaware State University, Dover, DE 19901, USA;
| | - Samuel N. Nahashon
- Department of Agriculture and Environmental Sciences, Tennessee State University, Nashville, TN 37209, USA;
| |
Collapse
|
7
|
Doranga S, Conway T. Nitrogen assimilation by E. coli in the mammalian intestine. mBio 2024; 15:e0002524. [PMID: 38380942 PMCID: PMC10936423 DOI: 10.1128/mbio.00025-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Nitrogen is an essential element for all living organisms, including Escherichia coli. Potential nitrogen sources are abundant in the intestine, but knowledge of those used specifically by E. coli to colonize remains limited. Here, we sought to determine the specific nitrogen sources used by E. coli to colonize the streptomycin-treated mouse intestine. We began by investigating whether nitrogen is limiting in the intestine. The NtrBC two-component system upregulates approximately 100 genes in response to nitrogen limitation. We showed that NtrBC is crucial for E. coli colonization, although most genes of the NtrBC regulon are not induced, which indicates that nitrogen is not limiting in the intestine. RNA-seq identified upregulated genes in colonized E. coli involved in transport and catabolism of seven amino acids, dipeptides and tripeptides, purines, pyrimidines, urea, and ethanolamine. Competitive colonization experiments revealed that L-serine, N-acetylneuraminic acid, N-acetylglucosamine, and di- and tripeptides serve as nitrogen sources for E. coli in the intestine. Furthermore, the colonization defect of a L-serine deaminase mutant was rescued by excess nitrogen in the drinking water but not by an excess of carbon and energy, demonstrating that L-serine serves primarily as a nitrogen source. Similar rescue experiments showed that N-acetylneuraminic acid serves as both a carbon and nitrogen source. To a minor extent, aspartate and ammonia also serve as nitrogen sources. Overall, these findings demonstrate that E. coli utilizes multiple nitrogen sources for successful colonization of the mouse intestine, the most important of which is L-serine. IMPORTANCE While much is known about the carbon and energy sources that are used by E. coli to colonize the mammalian intestine, very little is known about the sources of nitrogen. Interrogation of colonized E. coli by RNA-seq revealed that nitrogen is not limiting, indicating an abundance of nitrogen sources in the intestine. Pathways for assimilation of nitrogen from several amino acids, dipeptides and tripeptides, purines, pyrimidines, urea, and ethanolamine were induced in mice. Competitive colonization assays confirmed that mutants lacking catabolic pathways for L-serine, N-acetylneuraminic acid, N-acetylglucosamine, and di- and tripeptides had colonization defects. Rescue experiments in mice showed that L-serine serves primarily as a nitrogen source, whereas N-acetylneuraminic acid provides both carbon and nitrogen. Of the many nitrogen assimilation mutants tested, the largest colonization defect was for an L-serine deaminase mutant, which demonstrates L-serine is the most important nitrogen source for colonized E. coli.
Collapse
Affiliation(s)
- Sudhir Doranga
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tyrrell Conway
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
8
|
Shi J, Ma D, Gao S, Long F, Wang X, Pu X, Cannon RD, Han TL. Probiotic Escherichia coli Nissle 1917-derived outer membrane vesicles modulate the intestinal microbiome and host gut-liver metabolome in obese and diabetic mice. Front Microbiol 2023; 14:1219763. [PMID: 37649633 PMCID: PMC10465304 DOI: 10.3389/fmicb.2023.1219763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/26/2023] [Indexed: 09/01/2023] Open
Abstract
Introduction Obesity and diabetes are common chronic metabolic disorders which can cause an imbalance of the intestinal flora and gut-liver metabolism. Several studies have shown that probiotics, including Escherichia coli Nissle 1917 (EcN), promote microbial balance and metabolic health. However, there are no studies on how EcN outer membrane vesicles (EcN-OMVs) influence the intestinal microflora and affect the metabolic disorders of obesity and diabetes. Methods In this study, we evaluated the effects of EcN-OMVs on high-fat diet (HFD)-induced obesity and HFD + streptozotocin (STZ)-induced diabetes. Results EcN-OMVs could reduce body weight, decrease blood glucose, and increase plasma insulin in obese mice. Similarly, EcN-OMVs treatment could modify the ratio of Firmicutes/Bacteroidetes in the gut, elevate intestinal short-chain fatty acid (SCFA)-producing flora, and influence the SCFA content of the intestine. Furthermore, the intestinal metabolites ornithine and fumaric acid, hepatic ω-6 unsaturated fatty acids, and SCFAs were significantly increased after administering EcN-OMVs. Discussion Overall, this study showed that EcN-OMVs might act as post-biotic agents that could modulate gut-liver metabolism and ameliorate the pathophysiology of obesity and diabetes.
Collapse
Affiliation(s)
- Jun Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - DongXue Ma
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - ShanHu Gao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Fei Long
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Xin Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - XingYu Pu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Richard D. Cannon
- Department of Oral Sciences, Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| | - Ting-Li Han
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
He F, Jin X, Wang C, Hu J, Su S, Zhao L, Geng T, Zhao Y, Pan L, Bao N, Sun H. Lactobacillus rhamnosus GG ATCC53103 and Lactobacillus plantarum JL01 improved nitrogen metabolism in weaned piglets by regulating the intestinal flora structure and portal vein metabolites. Front Microbiol 2023; 14:1200594. [PMID: 37455717 PMCID: PMC10338925 DOI: 10.3389/fmicb.2023.1200594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
At present, most studies have shown that probiotics have a positive regulatory effect on the nutritional metabolism of the body, but the mechanism is still unclear. Here, 48 piglets were divided into four groups. The control group was not fed probiotics, the Lac group was fed L. Rhamnosus GG ATCC53103, the Rha group was fed L. Plantarum JL01, and the mix group was fed two types of probiotics. Nitrogen metabolism and mRNA levels of mTOR and S6K in skeletal muscle were observed in each group. Then, metagenome and non-targeted metabonomics were used to observe the changes of intestinal microorganisms and plasma metabolites in portal channels after probiotics feeding. Finally, we combined the results of omics analysis to reveal the mechanism of probiotics on nitrogen metabolism in weaned piglets. The results showed that L. Rhmnosus GG ATCC53103 and L. Plantarum JL01 increased nitrogen apparent digestibility, nitrogen deposition rate, and nitrogen utilization rate of weaned piglets (P < 0.05); the relative expression of mTOR and SK6 mRNA in skeletal muscle increased significantly (P < 0.05). When L. rhamnosus GG ATCC53103 and L. plantarum JL01 were combined, we found that Clostridium and Prevotella significantly increased in the jejunum (P < 0.05). The relative abundance of Lactobacillus, Ruminococcus, Streptococcus, and Prevotella in the ileum increased significantly (P < 0.05). Compared with the control group, L-Tryptophan, 3-Phosphonyloxypyruvate, cis-Aconitate, and Carbamoyl phosphate were significantly increased in the mixed group portal vein. The result of the combinatorial analysis showed that the significantly increased microorganisms could encode the enzyme genes for the synthesis of L-Tryptophan, 3-Phosphonooxypyruvate, cis-Aconitate, and Carbamoyl phosphate. In summary, our results demonstrated that L. Rhamnosus GG ATCC53103 and L. Plantarum JL01 could stimulate the expression of skeletal muscle protein synthesis genes of weaned piglets by modulating the structure of the gut microbiota and its metabolites, thereby improving nitrogen metabolism in weaned piglets.
Collapse
Affiliation(s)
- Feng He
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Xueying Jin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Shuai Su
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Lei Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Tingting Geng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Yuan Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Li Pan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Nan Bao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| |
Collapse
|
10
|
Abeyta MA, Horst EA, Mayorga EJ, Goetz BM, Al-Qaisi M, McCarthy CS, O'Neil MR, Dooley BC, Piantoni P, Schroeder GF, Baumgard LH. Effects of hindgut acidosis on metabolism, inflammation, and production in dairy cows consuming a standard lactation diet. J Dairy Sci 2023; 106:1429-1440. [PMID: 36460494 DOI: 10.3168/jds.2022-22303] [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: 05/16/2022] [Accepted: 08/21/2022] [Indexed: 11/30/2022]
Abstract
Postruminal intestinal barrier dysfunction caused by excessive hindgut fermentation may be a source of peripheral inflammation in dairy cattle. Therefore, the study objectives were to evaluate the effects of isolated hindgut acidosis on metabolism, inflammation, and production in lactating dairy cows. Five rumen-cannulated lactating Holstein cows (32.6 ± 7.2 kg/d of milk yield, 242 ± 108 d in milk; 642 ± 99 kg of body weight; 1.8 ± 1.0 parity) were enrolled in a study with 2 experimental periods (P). During P1 (4 d), cows were fed ad libitum a standard lactating cow diet (26% starch dry matter) and baseline data were collected. During P2 (7 d), all cows were fed the same diet ad libitum and abomasally infused with 4 kg/d of pure corn starch (1 kg of corn starch + 1.25 L of H2O/infusion at 0600, 1200, 1800, and 0000 h). Effects of time (hour relative to the first infusion or day) relative to P1 were evaluated using PROC MIXED in SAS (version 9.4; SAS Institute Inc.). Infusing starch markedly reduced fecal pH (5.84 vs. 6.76) and increased fecal starch (2.2 to 9.6% of dry matter) relative to baseline. During P2, milk yield, milk components, energy-corrected milk yield, and voluntary dry matter intake remained unchanged. At 14 h, plasma insulin and β-hydroxybutyrate increased (2.4-fold and 53%, respectively), whereas circulating glucose concentrations remained unaltered. Furthermore, blood urea nitrogen increased at 2 h (23%) before promptly decreasing below baseline at 14 h (13%). Nonesterified fatty acids tended to decrease from 2 to 26 h (40%). Circulating white blood cells and neutrophils increased on d 4 (36 and 73%, respectively) and somatic cell count increased on d 5 (4.8-fold). However, circulating serum amyloid A and lipopolysaccharide-binding protein concentrations were unaffected by starch infusions. Despite minor changes in postabsorptive energetics and leukocyte dynamics, abomasal starch infusions and the subsequent hindgut acidosis had little or no meaningful effects on biomarkers of immune activation or production variables.
Collapse
Affiliation(s)
- M A Abeyta
- Department of Animal Science, Iowa State University, Ames 50011
| | - E A Horst
- Department of Animal Science, Iowa State University, Ames 50011
| | - E J Mayorga
- Department of Animal Science, Iowa State University, Ames 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames 50011
| | - M Al-Qaisi
- Department of Animal Science, Iowa State University, Ames 50011
| | - C S McCarthy
- Department of Animal Science, Iowa State University, Ames 50011
| | - M R O'Neil
- Department of Animal Science, Iowa State University, Ames 50011
| | - B C Dooley
- Department of Animal Science, Iowa State University, Ames 50011
| | - P Piantoni
- Cargill Animal Nutrition Innovation Center, Elk River, MN 55330
| | - G F Schroeder
- Cargill Animal Nutrition Innovation Center, Elk River, MN 55330
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames 50011.
| |
Collapse
|
11
|
Li H, Li N, Lu Q, Yang J, Zhao J, Zhu Q, Yi S, Fu W, Luo T, Tang J, Zhang Y, Yang G, Liu Z, Xu J, Chen W, Zhu J. Chronic alcohol-induced dysbiosis of the gut microbiota and gut metabolites impairs sperm quality in mice. Front Microbiol 2022; 13:1042923. [PMID: 36532416 PMCID: PMC9751024 DOI: 10.3389/fmicb.2022.1042923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/04/2022] [Indexed: 08/23/2023] Open
Abstract
Studies have indicated that the ethanol exposure impairs the gut microbiota, At the same time, high levels of alcohol exposure damage sperm in mice. However, whether the gut microbiota is involved in mediating the effects of alcohol on sperm quality remains unclear. This study aimed to assess the effect of chronic alcohol consumption on intestinal microbiota in mice and analyze the potential pathophysiological effect of altered intestinal microbiota on sperm quality. We established a mouse model of chronic alcohol consumption by allowing male C57 mice to freely ingest 10% ethanol for 10 weeks, and collected the fecal microbiota of the male mice in the chronic drinking group (alcohol) and the control group (control) and transplanted the specimens into the transplant groups (the alcohol-fecal microbiota transplantation [FMT] group and the control-FMT group). Sperm quality was significantly decreased in the alcohol-FMT group compared with the control-FMT group. Gut microbiota analysis revealed that the abundance of 11 operational taxonomic units (OTUs) was altered in the alcohol-FMT group. Nontargeted metabolomics identified 105 differentially altered metabolites, which were mainly annotated to amino acids, lipids, glycerophosphoethanolamine, organic oxygenic compounds, organic acids and their derivatives, steroids, and flavonoids. In particular, the oxidative phosphorylation pathway, which is the key to spermatogenesis, was significantly enriched in the alcohol-FMT group. Moreover, compared with the control-FMT group, the alcohol-FMT group presented significantly higher serum endotoxin and inflammatory cytokine levels, with more pronounced T cell and macrophage infiltration in the intestinal lamina propria and elevated levels of testicular inflammatory cytokines. In addition, RNA sequencing showed significant differences in the expression of testis-related genes between the alcohol-FMT group and the control-FMT group. In particular, the expression of genes involved in gamete meiosis, testicular mitochondrial function, and the cell division cycle was significantly reduced in alcohol-FMT mice. In conclusion, these findings indicated that intestinal dysbiosis induced by chronic alcohol consumption may be an important factor contributing to impaired sperm quality. Chronic alcohol consumption induces intestinal dysbiosis, which then leads to metabolic disorders, elevated serum endotoxin and inflammatory cytokine levels, testicular inflammation, abnormal expression of related genes, and ultimately, impaired sperm quality. These findings are potentially useful for the treatment of male infertility.
Collapse
Affiliation(s)
- Hui Li
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Ningshan Li
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Qudong Lu
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jun Yang
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jiang Zhao
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Qiong Zhu
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Shanhong Yi
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Weihua Fu
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Tingting Luo
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jiawei Tang
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Yi Zhang
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guoliang Yang
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jie Xu
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Wei Chen
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jingzhen Zhu
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Zarei I, Koistinen VM, Kokla M, Klåvus A, Babu AF, Lehtonen M, Auriola S, Hanhineva K. Tissue-wide metabolomics reveals wide impact of gut microbiota on mice metabolite composition. Sci Rep 2022; 12:15018. [PMID: 36056162 PMCID: PMC9440220 DOI: 10.1038/s41598-022-19327-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 08/29/2022] [Indexed: 12/13/2022] Open
Abstract
The essential role of gut microbiota in health and disease is well recognized, but the biochemical details that underlie the beneficial impact remain largely undefined. To maintain its stability, microbiota participates in an interactive host-microbiota metabolic signaling, impacting metabolic phenotypes of the host. Dysbiosis of microbiota results in alteration of certain microbial and host metabolites. Identifying these markers could enhance early detection of certain diseases. We report LC-MS based non-targeted metabolic profiling that demonstrates a large effect of gut microbiota on mammalian tissue metabolites. It was hypothesized that gut microbiota influences the overall biochemistry of host metabolome and this effect is tissue-specific. Thirteen different tissues from germ-free (GF) and conventionally-raised (MPF) C57BL/6NTac mice were selected and their metabolic differences were analyzed. Our study demonstrated a large effect of microbiota on mammalian biochemistry at different tissues and resulted in statistically-significant modulation of metabolites from multiple metabolic pathways (p ≤ 0.05). Hundreds of molecular features were detected exclusively in one mouse group, with the majority of these being unique to specific tissue. A vast metabolic response of host to metabolites generated by the microbiota was observed, suggesting gut microbiota has a direct impact on host metabolism.
Collapse
Affiliation(s)
- Iman Zarei
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| | - Ville M Koistinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
- Food Chemistry and Food Development Unit, Department of Biochemistry, University of Turku, Itäinen Pitkäkatu 4, 20014, Turku, Finland
| | - Marietta Kokla
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Anton Klåvus
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Ambrin Farizah Babu
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
- LC-MS Metabolomics Center, Biocenter Kuopio, 70211, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
- LC-MS Metabolomics Center, Biocenter Kuopio, 70211, Kuopio, Finland
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Science, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
- Food Chemistry and Food Development Unit, Department of Biochemistry, University of Turku, Itäinen Pitkäkatu 4, 20014, Turku, Finland.
| |
Collapse
|
13
|
Dietary Alpha-Ketoglutarate Partially Abolishes Adverse Changes in the Small Intestine after Gastric Bypass Surgery in a Rat Model. Nutrients 2022; 14:nu14102062. [PMID: 35631203 PMCID: PMC9146360 DOI: 10.3390/nu14102062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/04/2022] [Accepted: 05/12/2022] [Indexed: 02/03/2023] Open
Abstract
Alpha-ketoglutarate (AKG) is one of the key metabolites that play a crucial role in cellular energy metabolism. Bariatric surgery is a life-saving procedure, but it carries many gastrointestinal side effects. The present study investigated the beneficial effects of dietary AKG on the structure, integrity, and absorption surface of the small intestine after bariatric surgery. Male 7-week-old Sprague Dowley rats underwent gastric bypass surgery, after which they received AKG, 0.2 g/kg body weight/day, administered in drinking water for 6 weeks. Changes in small intestinal morphology, including histomorphometric parameters of enteric plexuses, immunolocalization of claudin 3, MarvelD3, occludin and zonula ocludens 1 in the intestinal mucosa, and selected hormones, were evaluated. Proliferation, mucosal and submucosal thickness, number of intestinal villi and Paneth cells, and depth of crypts were increased; however, crypt activity, the absorption surface, the expression of claudin 3, MarvelD3, occludin and zonula ocludens 1 in the intestinal epithelium were decreased after gastric bypass surgery. Alpha-ketoglutarate supplementation partially improved intestinal structural parameters and epithelial integrity in rats undergoing this surgical procedure. Dietary AKG can abolish adverse functional changes in the intestinal mucosa, enteric nervous system, hormonal response, and maintenance of the intestinal barrier that occurred after gastric bypass surgery.
Collapse
|
14
|
Wu J, Zhang X, Tan Z, Jiao J. Distribution of free amino acids and mRNA expression of their corresponding transporters in the intestinal mucosa of goats feeding on a corn grain versus corn gluten diet. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:868-875. [PMID: 34218447 DOI: 10.1002/jsfa.11412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/10/2021] [Accepted: 07/04/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Intestinal amino acid (AA) chemosensing has been implicated in the regulation of AA absorption, nitrogen metabolism and hormone release, thereby playing an indispensable role in maintaining metabolic homeostasis in mammals. The objective of this experiment was to study the distribution of free AA and the expression of AA transporting related genes along the small and large intestines of Liuyang black goats, together with the effects of dietary corn grain replaced by dietary corn gluten feed (CGF). RESULTS The CGF replacement did not alter (P > 0.05) AA profiles and the expression of AA transporting related genes in the intestinal mucosa. Intriguingly, in terms of gut regions, the concentrations of aspartic acid and glutamic acid in the mucosa of ileum were remarkably less (P < 0.001) than those in the large intestine. Moreover, the concentrations of most cationic and neutral AAs shared the same distribution pattern, with the jejunum and ileum holding the greatest and least levels (P < 0.05), respectively. It was notable that the expression of both anionic and cationic AA transporters in the small intestine was exceedingly greater (P < 0.001) than those in the large intestine. As for transporters of neutral AA, system ASC, L, and A showed an extremely distinctive expression pattern. CONCLUSION The jejunum would be the primary site of transporting AA, while CGF substitution does not exert a disadvantageous influence on the AA chemosensing systems of the first-pass metabolism. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jian Wu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Xiaoli Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Zhiliang Tan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, P. R. China
| | - Jinzhen Jiao
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, P. R. China
| |
Collapse
|
15
|
Bergen WG. Pigs (Sus Scrofa) in Biomedical Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:335-343. [PMID: 34807450 DOI: 10.1007/978-3-030-85686-1_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Much of biomedical oriented research is conducted with animal models. Over the years, rodents (primarily rats and mice) have emerged as the preferred species for basic biochemistry, cell biology, physiology and nutrition studies. In the past, dogs have been used for the evaluation of dietary protein quality and other aspects of animal nitrogen metabolism and physiology, cardiovascular and endocrine research. At an increasing rate, pigs have also been used as a model species in biomedical research. Pigs are readily available in various mature sizes and genotypic/phenotypic traits, and there are many anatomic, nutritional and physiologic similarities between human beings and pigs. Many notable reviews summarizing the role of pigs in biomedical studies have already been published and these are cited below. The present review focuses on characteristics that make pigs an excellent biomedical animal model in particular in obesity, diabetes and cardiovascular research. To procure an animal model for obesity, irrespective of species used, these animals must be fed a dense caloric diet (high fat) to achieve an experimental working model within a reasonable period. This review also focuses on a putative role of gastrointestinal microbiota in obesity as obese animals exhibit a shift in the distribution of gastrointestinal microbial phyla from lean animals. But to date such results have not pinpointed a treatable cause for obesity. Sometimes, the choice of sampling sites for microbial assessment in many reports can be questioned as the microbial content and phyla distribution in easily collected fecal samples may differ from those obtained directly from the small intestine and upper colon. While pigs are still utilized in many countries for medical surgery practice, this has been discontinued in US medical schools.
Collapse
Affiliation(s)
- Werner G Bergen
- Department of Animal Sciences, Auburn University, AL, Auburn, 210 Upchurch Hall, 36854, USA.
| |
Collapse
|
16
|
Tuerhongjiang G, Guo M, Qiao X, Lou B, Wang C, Wu H, Wu Y, Yuan Z, She J. Interplay Between Gut Microbiota and Amino Acid Metabolism in Heart Failure. Front Cardiovasc Med 2021; 8:752241. [PMID: 34746265 PMCID: PMC8566708 DOI: 10.3389/fcvm.2021.752241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/07/2021] [Indexed: 11/14/2022] Open
Abstract
Heart failure (HF) is a complex clinical syndrome of which the incidence is on the rise worldwide. Cardiometabolic disorders are associated with the deterioration of cardiac function and progression of HF. Recently, there has been renewed interest in gut microbiota (GM) and its metabolites in the cardiovascular disease. HF-caused hypoperfusion could increase intestinal permeability, and a “leaky” bowel leads to bacterial translocation and make its metabolites more easily enter the circulation. Considerable evidence shows that the composition of microbiota and amino acids (AAs) has been altered in HF patients, and AAs could serve as a diagnostic and prognostic biomarker in HF. The findings indicate that the gut–amino acid–HF axis may play a key role in the progression of HF. In this paper, we focus on the interrelationship between the AA metabolism and GM alterations during the development of heart failure. We also discuss the potential prognostic and therapeutic value of the gut–amino acid–HF axis in the cortex of HF.
Collapse
Affiliation(s)
- Gulinigaer Tuerhongjiang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Manyun Guo
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Xiangrui Qiao
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Bowen Lou
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Chen Wang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Haoyu Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Yue Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Zuyi Yuan
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Jianqing She
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| |
Collapse
|
17
|
Liebe H, Liebe F, Sponder G, Hedtrich S, Stumpff F. Beyond Ca 2+ signalling: the role of TRPV3 in the transport of NH 4. Pflugers Arch 2021;473:1859-1884. [PMID: 34664138 PMCID: PMC8599221 DOI: 10.1007/s00424-021-02616-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Mutations of TRPV3 lead to severe dermal hyperkeratosis in Olmsted syndrome, but whether the mutants are trafficked to the cell membrane or not is controversial. Even less is known about TRPV3 function in intestinal epithelia, although research on ruminants and pigs suggests an involvement in the uptake of NH4+. It was the purpose of this study to measure the permeability of the human homologue (hTRPV3) to NH4+, to localize hTRPV3 in human skin equivalents, and to investigate trafficking of the Olmsted mutant G573S. Immunoblotting and immunostaining verified the successful expression of hTRPV3 in HEK-293 cells and Xenopus oocytes with trafficking to the cell membrane. Human skin equivalents showed distinct staining of the apical membrane of the top layer of keratinocytes with cytosolic staining in the middle layers. Experiments with pH-sensitive microelectrodes on Xenopus oocytes demonstrated that acidification by NH4+ was significantly greater when hTRPV3 was expressed. Single-channel measurements showed larger conductances in overexpressing Xenopus oocytes than in controls. In whole-cell experiments on HEK-293 cells, both enantiomers of menthol stimulated influx of NH4+ in hTRPV3 expressing cells, but not in controls. Expression of the mutant G573S greatly reduced cell viability with partial rescue via ruthenium red. Immunofluorescence confirmed cytosolic expression, with membrane staining observed in a very small number of cells. We suggest that expression of TRPV3 by epithelia may have implications not just for Ca2+ signalling, but also for nitrogen metabolism. Models suggesting how influx of NH4+ via TRPV3 might stimulate skin cornification or intestinal NH4+ transport are discussed.
Collapse
Affiliation(s)
- Hendrik Liebe
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163, Berlin, Germany.,Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Oertzenweg 19b, 14163, Berlin, Germany
| | - Franziska Liebe
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163, Berlin, Germany
| | - Gerhard Sponder
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163, Berlin, Germany
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Friederike Stumpff
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163, Berlin, Germany.
| |
Collapse
|
18
|
Méndez-Salazar EO, Martínez-Nava GA. Uric acid extrarenal excretion: the gut microbiome as an evident yet understated factor in gout development. Rheumatol Int 2021; 42:403-412. [PMID: 34586473 DOI: 10.1007/s00296-021-05007-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/19/2021] [Indexed: 12/19/2022]
Abstract
Humans do not produce uricase, an enzyme responsible for degrading uric acid. However, some bacteria residing in the gut can degrade one-third of the dietary and endogenous uric acid generated daily. New insights based on metagenomic and metabolomic approaches provide a new interest in exploring the involvement of gut microbiota in gout. Nevertheless, the exact mechanisms underlying this association are complex and have not been widely discussed. In this study, we aimed to review the evidence that suggests uric acid extrarenal excretion and gut microbiome are potential risk factors for developing gout. A literature search was performed in PubMed, Web of Science, and Google Scholar using several keywords, including "gut microbiome AND gout". A remarkable intestinal dysbiosis and shifts in abundance of certain bacterial taxa in gout patients have been consistently reported among different studies. Under this condition, bacteria might have developed adaptive mechanisms for de novo biosynthesis and salvage of purines, and thus, a concomitant alteration in uric acid metabolism. Moreover, gut microbiota can produce substrates that might cross the portal vein so the liver can generate de novo purinogenic amino acids, as well as uric acid. Therefore, the extrarenal excretion of uric acid needs to be considered as a factor in gout development. Nevertheless, further studies are needed to fully understand the role of gut microbiome in uric acid production and its extrarenal excretion, and to point out possible bacteria or bacterial enzymes that could be used as probiotic coadjutant treatment in gout patients.
Collapse
Affiliation(s)
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico.
| |
Collapse
|
19
|
Zhu P, Wong MKS, Lin X, Chan TF, Wong CKC, Lai KP, Tse WKF. Changes of the intestinal microbiota along the gut of Japanese Eel (Anguilla japonica). Lett Appl Microbiol 2021; 73:529-541. [PMID: 34265084 DOI: 10.1111/lam.13539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/21/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
Fish intestine contains different types of microbiomes, and bacteria are the dominant microbiota in fishes. Studies have identified various core gut bacteria in fishes. However, little is known about the composition and their relative functions of gut microbial community along the intestine. To explore this, the current study investigated the microbial community distribution along the gut in Anguilla japonica. By 16S rRNA gene sequencing, we profiled the gut microbiota in eel along the three regions (anterior intestine (AI), the middle intestine (MI) and the posterior intestine (PI)). Results suggested that the three regions did not have significant differences on the observed species and diversities. The cluster tree analysis showed that the bacteria community in MI was closer to PI than the AI. The dominant bacteria in AI were the Proteobacteria, in which the majority was graduated replaced by Bacteroidetes along the gut to PI region. Through PICRUSt analysis, shifts in the bacterial community along the gut were found to affect the genetic information processing pathways. Higher levels of translation and transcriptional pathway activities were found in MI and PI than in AI. The dominant bacterial species were different among the regions and contributed to various biological functions along the gut.
Collapse
Affiliation(s)
- P Zhu
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, Beibu Gulf University, Qinzhou, Guangxi, PR China
| | - M K-S Wong
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba, Japan
| | - X Lin
- School of Life Sciences, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - T F Chan
- School of Life Sciences, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - C K C Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guanzhou), The Hong Kong Baptist University, Kowloon, Hong Kong
| | - K P Lai
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, Beibu Gulf University, Qinzhou, Guangxi, PR China.,Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guanzhou), The Hong Kong Baptist University, Kowloon, Hong Kong.,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, PR China
| | - W K F Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
20
|
Lewton JR, Woodward AD, Moser RL, Thelen KM, Moeser AJ, Trottier NL, Tempelman RJ, Rozeboom DW. Effects of a multi-strain Bacillus subtilis-based direct-fed microbial on weanling pig growth performance and nutrient digestibility. Transl Anim Sci 2021; 5:txab058. [PMID: 34278233 PMCID: PMC8281103 DOI: 10.1093/tas/txab058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/17/2021] [Indexed: 12/31/2022] Open
Abstract
A study was conducted to evaluate the effects of a multi-strain Bacillus subtilis-based direct-fed microbial (DFM) on growth performance and apparent nutrient digestibility of nursery pigs. Eighty pigs, of equal number of barrows and gilts (initial body weight: 7.0 ± 0.60 kg), were weaned at 21 ± 1 d and randomly allotted to 1 of the 16 pens, with 5 pigs per pen. Two dietary treatments were implemented, a basal control (CON) and a control plus DFM (CDFM). Both diets were corn, soybean meal, and distillers dried grains based. Diets were fed for 42 d and growth performance measures were recorded weekly. On days 21 and 42 of the experiment, one pig per pen, with equal number of males and females, was randomly selected and euthanized. Digestibility of nitrogen (N), amino acids (AA), and energy were evaluated within the duodenum, jejunum, ileum, and ascending and distal colon. Relative to CON, CDFM tended to increase ADG during week 2 (P = 0.08) and significantly increased ADFI during week 2 (P = 0.04) and week 3 (P = 0.02). In addition, CDFM decreased the gain to feed ratio (G:F) during week 6 relative to CON (P = 0.04). Within the jejunum, pigs fed the DFM had greater digestibility of tryptophan (P = 0.04) and cysteine (P = 0.04) and tended to have greater digestibility of lysine (P = 0.07), methionine (P = 0.06), and threonine (P = 0.08), relative to CON. The content pH in the ascending colon did not differ between CDFM and CON. Compared with CON, apparent total tract digestibility (ATTD) of energy did not differ from CDFM, whereas ATTD of nitrogen of CDFM was lower (P = 0.05). The addition of a multi-strain B. subtilis-based DFM appears to impact growth performance, AA, and N digestibility depending upon the location in the gastrointestinal tract, with primary AA differences occurring within the mid-jejunum.
Collapse
Affiliation(s)
- Jaron R Lewton
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | | | | | - Kyan M Thelen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Adam J Moeser
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Nathalie L Trottier
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Robert J Tempelman
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Dale W Rozeboom
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
21
|
Liu H, Li Q, Zhao D, Zhang M, Jiang S, Li C. Changes in the structure and digestibility of myoglobin treated with sodium chloride. Food Chem 2021; 363:130284. [PMID: 34120050 DOI: 10.1016/j.foodchem.2021.130284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
Myoglobin is a protein not easily broken down by digestive enzymes due to its rigid structure. This study evaluated the structural characteristics of myoglobin under various sodium chloride treatments (0.4-0.8 mol/L for 5-10 h) and the impacts on its digestibility using spectroscopic and molecular dynamics simulation techniques. Myoglobin digestibility was 40% following pepsin digestion and 60% after being sequentially digested by pepsin and trypsin. The α-helix content of myoglobin did not change significantly following sodium chloride treatment but hydrophobic amino acids were exposed and the binding of phenylalanine targeted by some digestive enzymes became more stable, leading to the reduced digestibility.
Collapse
Affiliation(s)
- Hui Liu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qian Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Miao Zhang
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuai Jiang
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
22
|
Abdallah A, Elemba E, Zhong Q, Sun Z. Gastrointestinal Interaction between Dietary Amino Acids and Gut Microbiota: With Special Emphasis on Host Nutrition. Curr Protein Pept Sci 2021; 21:785-798. [PMID: 32048965 DOI: 10.2174/1389203721666200212095503] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 12/31/2022]
Abstract
The gastrointestinal tract (GIT) of humans and animals is host to a complex community of different microorganisms whose activities significantly influence host nutrition and health through enhanced metabolic capabilities, protection against pathogens, and regulation of the gastrointestinal development and immune system. New molecular technologies and concepts have revealed distinct interactions between the gut microbiota and dietary amino acids (AAs) especially in relation to AA metabolism and utilization in resident bacteria in the digestive tract, and these interactions may play significant roles in host nutrition and health as well as the efficiency of dietary AA supplementation. After the protein is digested and AAs and peptides are absorbed in the small intestine, significant levels of endogenous and exogenous nitrogenous compounds enter the large intestine through the ileocaecal junction. Once they move in the colonic lumen, these compounds are not markedly absorbed by the large intestinal mucosa, but undergo intense proteolysis by colonic microbiota leading to the release of peptides and AAs and result in the production of numerous bacterial metabolites such as ammonia, amines, short-chain fatty acids (SCFAs), branched-chain fatty acids (BCFAs), hydrogen sulfide, organic acids, and phenols. These metabolites influence various signaling pathways in epithelial cells, regulate the mucosal immune system in the host, and modulate gene expression of bacteria which results in the synthesis of enzymes associated with AA metabolism. This review aims to summarize the current literature relating to how the interactions between dietary amino acids and gut microbiota may promote host nutrition and health.
Collapse
Affiliation(s)
- Abedin Abdallah
- Key laboratory of Straw Biology and Utilization (The Ministry of Education), Key Lab of Animal Nutrition and Feed
Science, Key Lab of Animal Production, Product Quality and Security, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Evera Elemba
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Qingzhen Zhong
- Key laboratory of Straw Biology and Utilization (The Ministry of Education), Key Lab of Animal Nutrition and Feed
Science, Key Lab of Animal Production, Product Quality and Security, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zewei Sun
- Key laboratory of Straw Biology and Utilization (The Ministry of Education), Key Lab of Animal Nutrition and Feed
Science, Key Lab of Animal Production, Product Quality and Security, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
23
|
Ashaolu TJ, Ashaolu JO. Prebiotic peptides, their formation, fermentation in the gut, and health implications. Biotechnol Prog 2021; 37:e3142. [PMID: 33666376 DOI: 10.1002/btpr.3142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/20/2021] [Accepted: 01/30/2021] [Indexed: 12/19/2022]
Abstract
Prebiotics can be synthesized from sources other than dietary fibers, such as proteins. The proteins, when processed into peptides have healthful or deleterious effects on the host. Outside living systems, prebiotic peptides (PP) are formed via preformation of amino acids or related monomeric building blocks, resulting in nonenzymatic polymerization/ligation to produce peptides. Whereas, inside living systems like the human gut, many metabolic pathways are involved in PP production, and mostly involve host-microbiota interactions. The interplay is responsible for PP activities and their implications on host amino acid balance and metabolism. Similar to carbohydrates fermentation, PP will yield short chain fatty acids (SCFA), but also branched chain fatty acids (BCFAs), phenols, indole, hydrogen sulfide, amines, and ammonia, capable of biologically mediating molecular signals. This holistic review considers a brief description of prebiotics, and tracks down prebiotic peptides formation processes, interactions with gut microbes, and health outcomes.
Collapse
Affiliation(s)
- Tolulope Joshua Ashaolu
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam.,Faculty of Environmental and Chemical Engineering, Duy Tan University, Da Nang, Vietnam
| | - Joseph O Ashaolu
- International Health Programme, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
24
|
Pugliero S, Lima DY, Rodrigues AM, Bogsan CSB, Rogero MM, Punaro GR, Higa EMS. Kefir reduces nitrosative stress and upregulates Nrf2 in the kidney of diabetic rats. Int Dairy J 2021. [DOI: 10.1016/j.idairyj.2020.104909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
25
|
Lachica M, Rojas-Cano M, Lara L, Haro A, Fernández-Fígares I. Net portal appearance of proteinogenic amino acids in Iberian pigs fed betaine and conjugated linoleic acid supplemented diets. Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.114825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
26
|
Bergen WG. Amino Acids in Beef Cattle Nutrition and Production. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1285:29-42. [PMID: 33770401 DOI: 10.1007/978-3-030-54462-1_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Proteins have been recognized for a long time as an important dietary nutritional component for all animals. Most amino acids were isolated and characterized in the late nineteenth and early twentieth century. Initially dietary proteins were ranked high to low quality by growth and N balance studies. By the 1950s interest had shifted to studying the roles of individual amino acids in amino acid requirements by feeding studies with non-ruminants as rodents, poultry and pigs. The direct protein feeding approaches followed by measurements of nutritional outcomes were not possible however in ruminants (cattle and sheep). The development of measuring free amino acids by ion exchange chromatography enabled plasma amino acid analysis. It was thought that plasma amino acid profiles were useful in nutritional studies on proteins and amino acids. With non-ruminants, nutritional interpretations of plasma amino acid studies were possible. Unfortunately with beef cattle, protein/amino acid nutritional adequacy or requirements could not be routinely determined with plasma amino acid studies. In dairy cows, however, much valuable understanding was gained from amino acid studies. Concurrently, others studied amino acid transport in ruminant small intestines, the role of peptides in ruminant N metabolism, amino acid catabolism (in the animal) with emphasis on branched-chain amino acid catabolism. In addition, workable methodologies for studying protein turnover in ruminants were developed. By the 1990s, nutritionists could still not determine amino acid requirements with empirical experimental studies in beef cattle. Instead, computer software (expert systems) based on the accumulated knowledge in animal and ruminal amino acids, energy metabolism and protein production were realized and revised frequently. With these tools, the amino acid requirements, daily energy needs, ruminal and total gastrointestinal tract digestion and performance of growing beef cattle could be predicted.
Collapse
Affiliation(s)
- Werner G Bergen
- Department of Animal Sciences, Auburn University, Auburn, AL, USA.
| |
Collapse
|
27
|
Herring CM, Bazer FW, Wu G. Amino Acid Nutrition for Optimum Growth, Development, Reproduction, and Health of Zoo Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1285:233-253. [PMID: 33770410 DOI: 10.1007/978-3-030-54462-1_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteins are large polymers of amino acids (AAs) linked via peptide bonds, and major components for the growth and development of tissues in zoo animals (including mammals, birds, and fish). The proteinogenic AAs are alanine, arginine, aspartate, asparagine, cysteine, glutamate, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine. Except for glycine, they are all present in the L-isoform. Some carnivores may also need taurine (a nonproteinogenic AA) in their diet. Adequate dietary intakes of AAs are necessary for the growth, development, reproduction, health and longevity of zoo animals. Extensive research has established dietary nutrient requirements for humans, domestic livestock and companion animals. However, this is not true for many exotic or endangered species found in zoos due to the obstacles that accompany working with these species. Information on diets and nutrient profiles of free-ranging animals is needed. Even with adequate dietary intake of crude protein, dietary AAs may still be unbalanced, which can lead to nutrition-related diseases and disorders commonly observed in captive zoo species, such as dilated cardiomyopathy, urolithiasis, gut dysbiosis, and hormonal imbalances. There are differences in AA metabolism among carnivores, herbivores and omnivores. It is imperative to consider these idiosyncrasies when formulating diets based on established nutritional requirements of domestic species. With optimal health, populations of zoo animals will have a vastly greater chance of thriving in captivity. For endangered species especially, maintaining stable captive populations is crucial for conservation. Thus, adequate provision of AAs in diets plays a crucial role in the management, sustainability and expansion of healthy zoo animals.
Collapse
Affiliation(s)
- Cassandra M Herring
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
28
|
Yu M, Li Z, Wang G, Cui Y, Rong T, Tian Z, Liu Z, Li J, Chen W, Ma X. Dietary supplementation with citrus extract alters the plasma parameters, circulating amino acid profiles and gene expression of small intestinal nutrient transporters in Chinese yellow-feathered broilers. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:5126-5135. [PMID: 32469078 DOI: 10.1002/jsfa.10525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/11/2020] [Accepted: 05/29/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND This study evaluated the effects of citrus extract (CE) on growth performance, plasma amino acid (AA) profiles, intestinal development and small intestine AA and peptide transporter expression levels in broilers. A total of 540 one-day-old yellow-feathered broilers were fed a basal diet without any antibiotic (control group), or a basal diet containing 10 mg kg-1 zinc bacitracin (antibiotic group), or a basal diet supplemented with 10 mg kg-1 CE (CE group). After 63 days of feeding, two broilers per pen were slaughtered to collect tissues for further analysis. RESULTS Results showed that CE increased (P < 0.05) the final body weight and average daily gain from day 1 to 63, and decreased (P < 0.05) the feed/gain ratio from day 1 to 63. Dietary CE supplementation increased (P < 0.05) plasma total protein, albumin and glucose concentration, and decreased (P < 0.05) urea concentration. CE supplementation increased (P < 0.05) the villus height in the ileum and the villus height/crypt depth in the jejunum and ileum, but decreased (P < 0.05) the crypt depth in the jejunum and ileum. CE supplementation increased (P < 0.05) most plasma essential AA concentrations. Additionally, CE supplementation upregulated (P < 0.05) ASCT1, b0,+ AT, B0 AT1, EAAT3, rBAT, y+ LAT2 and PepT1 expression in the jejunum, and b0,+ AT, EAAT3, rBAT, y+ LAT2, CAT1 and PepT1 in the ileum. CONCLUSIONS Collectively, our results indicated that CE supplementation promotes intestinal physiological absorption of AAs by upregulating gene expression of small intestinal key AA and peptide transporters, thereby enhancing the growth performance of broilers. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Miao Yu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhenming Li
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Gang Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Yiyan Cui
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Ting Rong
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhimei Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zhichang Liu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Jiazhou Li
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Weidong Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Xianyong Ma
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences; State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs; Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition; Guangdong Engineering Technology Research Center of animal Meat quality and Safety Control and Evaluation; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
29
|
Sun M, Zhao J, Wang X, Jiao H, Lin H. Use of encapsulated L-lysine-HCl and DL-methionine improves postprandial amino acid balance in laying hens. J Anim Sci 2020; 98:skaa315. [PMID: 32954399 PMCID: PMC7759752 DOI: 10.1093/jas/skaa315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/17/2020] [Indexed: 11/12/2022] Open
Abstract
The supplementation of dietary limiting amino acids (AA) with crystalline AA makes the use of low-protein diets an option in poultry production. The differing absorption rates of crystalline and protein-bound AA may lead to temporally imbalanced AA in the postabsorptive period. In this study, two experiments were conducted to evaluate the effect of encapsulated L-lysine-HCl (L-Lys-HCl) and DL-methionine (DL-Met) on the laying performance of hens. In exp. 1, a total of 135 forty-seven-wk-old Hy-Line Brown hens were subjected to three dietary treatments for 8 wk: basal diet supplemented with 0.14% L-Lys-HCl and 0.17% DL-Met to satisfy the NRC (1994) total Lys and Met recommendation (control) and basal diet supplemented with encapsulated L-Lys-HCl and DL-Met at the levels of 60% (60CLM, 0.084% L-Lys-HCl and 0.102% DL-Met) or 80% of control (80CLM, 0.112% L-Lys-HCl and 0.136% DL-Met), respectively. In exp. 2, 24 fifty-five-wk-old Hy-Line Brown hens were individually reared in cages and subjected to the same treatments as in exp. 1. The plasma concentrations of free AA and nitrogen metabolites were measured 2, 4, and 6 h after fed. The results showed that dietary AA treatment had no significant influence on body weight (BW), feed intake, laying rate, egg weight, egg mass, or feed efficiency. The expression levels of AA transporters CAT-1, y+LAT1, b0,+AT, B0AT, rBAT, EAAT3, and PepT1 in the duodenum, jejunum, and ileum were not influenced (P > 0.05) by dietary treatment. There was an interaction of dietary AA treatment and time (P < 0.05) and the 80CLM hens exhibited higher concentrations of Lys (P < 0.05) than the controls at 2-h time point. In contrast, plasma Met concentration was not influenced (P > 0.05), while Cys was reduced in the 60CLM hens at every time point. The 80CLM hens had higher taurine concentrations than those receiving the control diet at every postprandial time point. In conclusion, these findings demonstrate that by using encapsulated form, the supplemental levels of synthetic L-Lys-HCl and DL-Met can be effectively reduced by approximately 20% with no negative effect on laying performance. The result suggests that encapsulated Lys and Met may ameliorate the postabsorptive AA balance and contribute to the reduced dietary AA supplemental levels.
Collapse
Affiliation(s)
- Mingfa Sun
- Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Jingpeng Zhao
- Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaojuan Wang
- Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Hongchao Jiao
- Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Hai Lin
- Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
30
|
Sun M, Jiao H, Wang X, Uyanga VA, Zhao J, Lin H. Encapsulated crystalline lysine and DL-methionine have higher efficiency than the crystalline form in broilers. Poult Sci 2020; 99:6914-6924. [PMID: 33248607 PMCID: PMC7704974 DOI: 10.1016/j.psj.2020.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/06/2020] [Accepted: 09/07/2020] [Indexed: 11/28/2022] Open
Abstract
Crystalline amino acids (AAs) exhibit high nutritional values when supplemented AA-deficient diets. However, the AAs in crystalline form in the diet are absorbed quickly than protein-bound AAs, which may take an effect on AA utilization efficiency. In this study, 2 experiments were conducted to investigate the effect of encapsulated lysine-HCl (Lys) and DL-methionine (DL-Met) on the growth performance of broiler chickens. In experiment 1, a total of 432 one-day-old male Arbor Acres broilers were subjected to 3 dietary treatments (27 pens; 16 birds per pen) for 42 d. The control group was basal diets supplemented with crystalline Lys and DL-Met, and treatment groups had basal diets supplemented with encapsulated Lys and DL-Met at the levels of 80% and 60% of control diets (80CLM, 60CLM), respectively. The growth performance, intestinal development, and transcription of AA transporters were determined. In experiment 2, 24 broiler chickens were subjected to the same treatments as in experiment 1. The plasma concentrations of free AAs were measured 0, 2, 4, and 6 h after feeding. The results showed that 80CLM treatment had no significant influence on production performance, carcass characteristics, and plasma free AAs content during the experiment compared with the control group (P > 0.05). In addition, the 80CLM group moderately enhanced gut morphology development and increased AAs' absorption capacity. However, broilers fed the 60CLM diet had lower production performance and breast muscle weight than the control group (P < 0.05), but increased villi height and B0AT mRNA expression level (P < 0.05). At h 4 after feeding, the 60CLM broilers exhibited higher concentration of Ala, Cys, and total dispensable AAs than the control group (P < 0.05). In conclusion, the result suggests that the supplemental levels of crystalline Lys and DL-Met can be effectively saved approximately for 20% by using the encapsulated form in broilers, with improvements to AAs utilization efficiency, while posing no detrimental effects on production performance. Encapsulated Lys and DL-Met would have greater potential for application when replacing crystalline AAs in broiler chickens.
Collapse
Affiliation(s)
- Mingfa Sun
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Hongchao Jiao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Xiaojuan Wang
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Victoria A Uyanga
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China
| | - Jingpeng Zhao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China.
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control and Prevention, Taian City, Shandong Province, 271018, China.
| |
Collapse
|
31
|
The Metabolic Effects of Pre-probiotic Supplementation After Roux-en-Y Gastric Bypass (RYGB) Surgery: a Prospective, Randomized Controlled Study. Obes Surg 2020; 31:215-223. [PMID: 32803709 DOI: 10.1007/s11695-020-04894-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Following Roux-en-Y gastric bypass (RYGB), positive alterations are observed in gut microbiota and intestinal peptides. Previous studies demonstrated similar alterations observed in cases when pre-probiotics are used without surgery. The aim of this trial was to evaluate the effectiveness of early use of pre-probiotics after RYGB. MATERIAL AND METHODS The operation and follow-up of the patients were performed at Istanbul University Medical Faculty. Thirty-two patients who had undergone RYGB were randomized to pre-probiotic group (PreProBG, n = 16; 200 g/day yogurt plus 10 g/day inulin+oligofructose) and probiotic group (ProBG, n = 16; 200 g/day yogurt only) for 6 months. Blood samples (glucose, insulin, A1c, GLP-1, PYY), anthropometric measurements, and appetite ratings have been evaluated at baseline and 3 (m3) and 6 (m6) months after RYGB. RESULTS Initial anthropometric measurements and appetite ratings decreased significantly after surgery and there were no significant differences between the groups. The decrease of area under the curve(insulin) was less and has a positive correlation with the changes in anthropometric measurements in PreProBG. GLP-1 and PYY which increased dramatically after surgery in all patients were higher in PreProBG. But this increase had a negative correlation with the changes in anthropometric measurements during the study. CONCLUSION Increased insulin, GLP-1, and PYY secretion was more enhanced by pre-probiotic use in early postoperative period. But this increase not only in anthropometric measurements but also in appetite ratings affects negatively, contrary to expectations. In summary, it should be investigated with new studies that use of pre-probiotics in the late postoperative period may be more effective in patients with weak insulin and incretin response and therefore insufficient weight loss. Trial Registration clinicaltrials.gov Identifier: NCT03517345.
Collapse
|
32
|
Fukizawa S, Yamashita M, Fujisaka S, Tobe K, Nonaka Y, Murayama N. Isoxanthohumol, a hop-derived flavonoid, alters the metabolomics profile of mouse feces. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2020; 39:100-108. [PMID: 32775127 PMCID: PMC7392914 DOI: 10.12938/bmfh.2019-045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022]
Abstract
The aim of this study was to verify the effect of treatment with isoxanthohumol (IX) on the metabolomics profile of mouse feces to explore the host-intestinal bacterial interactions at the molecular level. The fecal contents of several amino acids in the high-fat diet (HFD) + 0.1% IX group (treated with IX mixed in diets for 12 weeks) were significantly lower than in the HFD group. The fecal contents of the secondary bile acid deoxycholic acid (DCA) in the HFD + 180 mg/kg IX group (orally treated with IX for 8 weeks) were significantly lower than in the HFD group; the values in the HFD group were higher than those in the normal diet (ND) group. Administration of IX changed the fecal metabolomics profile. For some metabolites, IX normalized HFD-induced fluctuations.
Collapse
Affiliation(s)
- Shinya Fukizawa
- Research Institute, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Mai Yamashita
- Research Institute, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
| | - Yuji Nonaka
- Research Institute, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Norihito Murayama
- Research Institute, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| |
Collapse
|
33
|
Picca A, Calvani R, Cesari M, Landi F, Bernabei R, Coelho-Júnior HJ, Marzetti E. Biomarkers of Physical Frailty and Sarcopenia: Coming up to the Place? Int J Mol Sci 2020; 21:E5635. [PMID: 32781619 PMCID: PMC7460617 DOI: 10.3390/ijms21165635] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Physical frailty and sarcopenia (PF&S) recapitulates all the hallmarks of aging and has become a focus in geroscience. Factors spanning muscle-specific processes (e.g., mitochondrial dysfunction in skeletal myocytes) to systemic changes (e.g., inflammation and amino acid dysmetabolism) have been pinpointed as possible contributors to PF&S pathophysiology. However, the search for PF&S biomarkers allowing the early identification and tracking of the condition over time is ongoing. This is mainly due to the phenotypic heterogeneity of PF&S, its unclear pathophysiology, and the frequent superimposition of other age-related conditions. Hence, presently, the identification of PF&S relies upon clinical, functional, and imaging parameters. The adoption of multi-marker approaches (combined with multivariate modeling) has shown great potential for addressing the complexity of PF&S pathophysiology and identifying candidate biological markers. Well-designed longitudinal studies are necessary for the incorporation of reliable biomarkers into clinical practice and for unveiling novel targets that are amenable to interventions.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, Università di Milano, 20122 Milan, Italy;
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Department of Geriatric and Orthopedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Roberto Bernabei
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Department of Geriatric and Orthopedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Hélio José Coelho-Júnior
- Department of Geriatric and Orthopedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Department of Geriatric and Orthopedic Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
34
|
Impact of Protein Intake in Older Adults with Sarcopenia and Obesity: A Gut Microbiota Perspective. Nutrients 2020; 12:nu12082285. [PMID: 32751533 PMCID: PMC7468805 DOI: 10.3390/nu12082285] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
The continuous population increase of older adults with metabolic diseases may contribute to increased prevalence of sarcopenia and obesity and requires advocacy of optimal nutrition treatments to combat their deleterious outcomes. Sarcopenic obesity, characterized by age-induced skeletal-muscle atrophy and increased adiposity, may accelerate functional decline and increase the risk of disability and mortality. In this review, we explore the influence of dietary protein on the gut microbiome and its impact on sarcopenia and obesity. Given the associations between red meat proteins and altered gut microbiota, a combination of plant and animal-based proteins are deemed favorable for gut microbiota eubiosis and muscle-protein synthesis. Additionally, high-protein diets with elevated essential amino-acid concentrations, alongside increased dietary fiber intake, may promote gut microbiota eubiosis, given the metabolic effects derived from short-chain fatty-acid and branched-chain fatty-acid production. In conclusion, a greater abundance of specific gut bacteria associated with increased satiation, protein synthesis, and overall metabolic health may be driven by protein and fiber consumption. This could counteract the development of sarcopenia and obesity and, therefore, represent a novel approach for dietary recommendations based on the gut microbiota profile. However, more human trials utilizing advanced metabolomic techniques to investigate the microbiome and its relationship with macronutrient intake, especially protein, are warranted.
Collapse
|
35
|
Jian X, Zhu Y, Ouyang J, Wang Y, Lei Q, Xia J, Guan Y, Zhang J, Guo J, He Y, Wang J, Li J, Lin J, Su M, Li G, Wu M, Qiu L, Xiang J, Xie L, Jia W, Zhou W. Alterations of gut microbiome accelerate multiple myeloma progression by increasing the relative abundances of nitrogen-recycling bacteria. MICROBIOME 2020; 8:74. [PMID: 32466801 PMCID: PMC7257554 DOI: 10.1186/s40168-020-00854-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/04/2020] [Indexed: 05/05/2023]
Abstract
BACKGROUND Gut microbiome alterations are closely related to human health and linked to a variety of diseases. Although great efforts have been made to understand the risk factors for multiple myeloma (MM), little is known about the role of the gut microbiome and alterations of its metabolic functions in the development of MM. RESULTS Here, in a cohort of newly diagnosed patients with MM and healthy controls (HCs), significant differences in metagenomic composition were discovered, for the first time, with higher bacterial diversity in MM. Specifically, nitrogen-recycling bacteria such as Klebsiella and Streptococcus were significantly enriched in MM. Also, the bacteria enriched in MM were significantly correlated with the host metabolome, suggesting strong metabolic interactions between microbes and the host. In addition, the MM-enriched bacteria likely result from the regulation of urea nitrogen accumulated during MM progression. Furthermore, by performing fecal microbiota transplantation (FMT) into 5TGM1 mice, we proposed a mechanistic explanation for the interaction between MM-enriched bacteria and MM progression via recycling urea nitrogen. Further experiments validated that Klebsiella pneumoniae promoted MM progression via de novo synthesis of glutamine in mice and that the mice fed with glutamine-deficient diet exhibited slower MM progression. CONCLUSIONS Overall, our findings unveil a novel function of the altered gut microbiome in accelerating the malignant progression of MM and open new avenues for novel treatment strategies via manipulation of the intestinal microbiota of MM patients. Video abstract.
Collapse
Affiliation(s)
- Xingxing Jian
- State Key Laboratory of Experimental Hematology, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, China
| | - Yinghong Zhu
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Jian Ouyang
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, China
| | - Yihui Wang
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Qian Lei
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Jiliang Xia
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yongjun Guan
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Jingyu Zhang
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Jiaojiao Guo
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yanjuan He
- State Key Laboratory of Experimental Hematology, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinuo Wang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingchao Lin
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Mingming Su
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Guancheng Li
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Juanjuan Xiang
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, China
| | - Wei Jia
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Wen Zhou
- State Key Laboratory of Experimental Hematology, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, China-Africa Research Center of Infectious Deseases, Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
36
|
Xie Y, Wang C, Zhao D, Zhou G, Li C. Processing Method Altered Mouse Intestinal Morphology and Microbial Composition by Affecting Digestion of Meat Proteins. Front Microbiol 2020; 11:511. [PMID: 32322243 PMCID: PMC7156556 DOI: 10.3389/fmicb.2020.00511] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Our previous study showed that the intake of meat proteins dynamically affected fecal microbial composition. However, the digestion of processed meat proteins in vivo and its relationship with gut microbiota and host remain unclear. In this study, we collected cecal contents and intestinal tissue from the mice fed with casein, soybean protein (SP), and four processed pork proteins for 8 months, and analyzed the amino acid (AA) files, cecum microbial composition and metabolites, and intestinal morphology. Dry-cured pork protein and stewed pork protein (SPP) groups had significantly higher total AA content in gut content than the other groups, but the content of the SPP group was relatively lower in the serum. The microbial composition of the processed meat protein groups differed from the casein or SP group, which is consistent with changes in AA composition. Emulsion sausage protein and SP diets upregulated the microbial AA metabolism, energy metabolism, signaling molecules and interaction, translation, and digestive system function but downregulated the microbial membrane transport, signal transduction and cell motility function compared to the casein diet. The SPP diets increased concentrations of acetate, propionate, butyrate, and isovalerate by specific gut microbes, but it decreased the relative abundance of Akkermansia. Moreover, the mice fed SP diet had relatively lower crypt depth, higher villus height and V/C ratio in duodenum, with the longer small intestines and the heavier cecum than other diets. These results suggested that processing methods altered bioavailability of meat proteins, which affected the intestinal morphology and the cecum microbial composition and function.
Collapse
Affiliation(s)
- Yunting Xie
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MARA, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chong Wang
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MARA, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MARA, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MARA, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
- International Joint Laboratory of Animal Health and Food Safety, MOE, Nanjing Agricultural University, Nanjing, China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MARA, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
- International Joint Laboratory of Animal Health and Food Safety, MOE, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
37
|
Yang J, Tan B, He Q, Yin Y, Wu G, Kong X. Dynamic changes in circulating levels of metabolites in the portal-drained viscera of finishing pigs receiving acute administration of l-arginine. J Anim Physiol Anim Nutr (Berl) 2020; 104:1424-1431. [PMID: 32227548 DOI: 10.1111/jpn.13350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 11/30/2022]
Abstract
In this study, we examined the effects of acute intravenous administration of l-arginine on circulating levels of metabolites in the portal-drained viscera (PDV) of 12 barrows surgically fitted with chronic catheters in the portal vein. At day 14 post-surgery, the pigs were fasted for 12 hr and then randomly allocated to one of three groups to receive administration of normal saline, l-alanine [103 mg/kg body weight (BW), isonitrogenous control] or l-arginine-HCl (61 mg/kg BW), via the portal vein. Blood samples were obtained from the carotid artery before and at 30-min intervals for 5 hr after the administration of saline or amino acid in order to determine metabolic profiles. The results showed that, compared with the saline treatment, arginine infusion increased plasma concentrations of insulin-like growth factor-I, arginine and cystine in the portal vein plasma, whereas plasma concentrations of threonine, serine, leucine and methionine were reduced. These findings indicate that increasing arginine concentrations in the portal vein alters the metabolic profile in swine, an established animal model for studying human nutrition and metabolism.
Collapse
Affiliation(s)
- Jianying Yang
- Hunan Provincial Key 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.,College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Bi'e Tan
- Hunan Provincial Key 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
| | - Qinghua He
- Hunan Provincial Key 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
| | - Yulong Yin
- Hunan Provincial Key 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
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Xiangfeng Kong
- Hunan Provincial Key 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
| |
Collapse
|
38
|
Qiao J, Li H, Li Y. Dietary Clostridium butyricum supplementation modifies significantly the liver transcriptomic profile in weaned piglets. J Anim Physiol Anim Nutr (Berl) 2020; 104:1410-1423. [PMID: 32207194 DOI: 10.1111/jpn.13326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/20/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022]
Abstract
The addition of probiotics in swine nutrition is known to positively influence both health and growth. The current study investigates differences in the hepatic transcriptome profiles between weaned piglets supplemented with Clostridium butyricum (C. butyricum) and control animals that received no probiotic. The liver is an important metabolic organ that plays a critical role in oxidizing triglycerides for energy production, lipid synthesis and degradation, as well as immune regulation in animals. RNA-Seq analysis was carried out on total RNA harvested from the liver of piglets fed with (n = 3) or without (n = 3) 5 × 105 C. butyricum CFU/g. Compared to the control piglets, 588 of the genes examined (352 up-regulated and 236 down-regulated) were significantly differentially expressed at a fold change > 2 and p < .05 in animals fed with C. butyricum. Quantitative real-time reverse transcription PCR (qRT-PCR) analysis was further used to validate the microarray expression results for 28 genes tested. The functional annotation analyses revealed several genes, processes and pathways with putative involvement in piglet growth and performance. Feeding swine with 5 × 105 C. butyricum CFU/g appears to reinforce their immune status as well as foster the cell cycle and improve the metabolism of carbohydrates, lipids and amino acids. This study provides valuable information about the expression profiles of mRNAs in piglet liver and in-depth functional investigations of these mRNAs that could provide new insights into the molecular networks of growth, immune responses and nutrient metabolism in the porcine liver.
Collapse
Affiliation(s)
- Jiayun Qiao
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Haihua Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yupeng Li
- Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| |
Collapse
|
39
|
Kerr BJ, Curry SM, Ramirez BC. Lack of interactive effects between diet composition and acid addition with drying method on amino acid digestibility values in porcine ileal digesta. J Anim Sci 2020; 98:skaa026. [PMID: 31993632 PMCID: PMC7038183 DOI: 10.1093/jas/skaa026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/24/2020] [Indexed: 11/13/2022] Open
Abstract
Two experiments were conducted to determine the effect of oven drying (OD) or freeze drying (FD) on apparent ileal digestibility (AID) of AA in diets fed to pigs. In experiment 1, 15 barrows (88.4 ± 6.4 kg) were allotted to either a corn starch-soybean meal (CST), potato starch-soybean meal (PST), or corn-soybean meal (CSBM) diet, over 2 collection periods. Following collection, samples were pooled within pig and subdivided into either OD or FD, resulting in 10 observations per diet by drying-method combination. In experiment 2, 11 barrows (63.3 ± 3.8 kg) were fed a CST diet, and, following collection, samples were pooled within pig and subdivided and either adjusted to pH 4 or remain unadjusted. Subsets of these samples were then subdivided to be either FD or OD, resulting in 11 observations per pH level by drying-method combination. OD was accomplished by drying samples in a forced air oven at either 100 °C (experiment 1) or 75 °C (experiment 2). In experiment 1, there was no diet type by drying-method interactions noted for any of the AA (P > 0.10).OD resulted in a higher AID of AA compared with samples which were FD (P ≤ 0.10), for all AA except for Gly and Tyr. Averaged across all AA, AID of AA was 3.3% greater if the sample was OD compared with FD. Differences in AID of AA among the 3 diets were noted for all AA (P ≤ 0.07), except for Cys (P = 0.33), due to the fact that CST and PST diets only contained soybean meal (SBM) as an AA-providing ingredient while the CSBM diet contained both corn and SBM. Pigs fed the PST diet had greater SID for all AA compared with pigs fed the CST diet (P ≤ 0.05), except for His, Lys, Cys, and Glu. In experiment 2, there were no pH-adjustment by drying-method interactions noted on AID for any of the AA (P > 0.10). Adjusting ileal digesta to a pH of 4.0 had little effect on AID for most of the AA, except for a lowered AID of Arg, His, Lys, Trp, and Ser (P ≤ 0.10). OD resulted in a higher AID for all AA (P ≤ 0.09) except for Ile, Thr, Val, Ala, Asp, Glu, and Gly. Averaged across all AA, the increase in AID of AA was 1.7% greater if the sample was OD compared with FD. On average, OD of ileal digesta resulted in a 2.5% greater estimate of AID of AA compared with samples that were FD, and was not diet-, pH-, or AA-dependent. Because the majority of the data on AA digestibility are based on FD, a bias factor may be necessary to adjust AA digestibility data obtained on an OD-basis relative to an FD-basis for use in feed formulation.
Collapse
Affiliation(s)
- Brian J Kerr
- U.S. Department of Agriculture-Agriculture Research Service, Ames, IA
| | - Shelby M Curry
- Oak Ridge Institute for Science and Education, Oak Ridge, TN
| | - Brett C Ramirez
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, IA
| |
Collapse
|
40
|
Yu M, Mu C, Zhang C, Yang Y, Su Y, Zhu W. Long-term effect of early antibiotic exposure on amino acid profiles and gene expression of transporters and receptors in the small intestinal mucosa of growing pigs with different dietary protein levels. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:235-244. [PMID: 31512251 DOI: 10.1002/jsfa.10028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND This study evaluated the effects of early antibiotic exposure (EAE) on subsequent amino acid (AA) profiles and small intestinal AA transporter and receptor expression level in pigs with different dietary crude protein (CP) levels. Eighteen litters of piglets were fed creep feed diets, either with or without antibiotics while with sow on day 7. The pigs were weaned at day 23 and fed the same diets until day 42, when random pigs within each group were offered a normal- or low-CP diet, thereby creating four groups. On day 120, the pigs were euthanized, and jejunal and ileal mucosa and digesta were collected for gene-expression and AA-concentration analysis. RESULTS With the normal-CP diet, EAE increased (P < 0.05) the concentrations of six essential amino acids (EAA) and three non-essential amino acids (NEAA) in serum, four EAAs and four NEAAs in jejunal mucosa, one EAA and two NEAAs in ileal mucosa, five EAAs and three NEAAs in jejunal digesta, and three EAAs and two NEAAs in ileal digesta. Early antibiotic exposure upregulated (P < 0.05) CAT1, ASCT2, ATB0,+ , CaSR, T1R1, and T1R3 expression in the jejunum, downregulated PepT1 expression with a normal-CP diet. It upregulated (P < 0.05) the expressions of CAT1, ATB0,+ , ATP1A1, and T1R3 in the ileum with a normal-CP diet. CONCLUSION These results suggest that EAE has long-term effects on AA profiles, mainly in the jejunum and serum, by increasing AA transporter expression in the intestine, and that these effects may be influenced by dietary CP levels. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Miao Yu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture Guangdong Public Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| | - Chunlong Mu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chuanjian Zhang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuxiang Yang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yong Su
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
41
|
Chen Y, Dinges MM, Green A, Cramer SE, Larive CK, Lytle C. Absorptive transport of amino acids by the rat colon. Am J Physiol Gastrointest Liver Physiol 2020; 318:G189-G202. [PMID: 31760764 PMCID: PMC6985843 DOI: 10.1152/ajpgi.00277.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The capacity of the colon to absorb microbially produced amino acids (AAs) and the underlying mechanisms of AA transport are incompletely defined. We measured the profile of 16 fecal AAs along the rat ceco-colonic axis and compared unidirectional absorptive AA fluxes across mucosal tissues isolated from the rat jejunum, cecum, and proximal colon using an Ussing chamber approach, in conjunction with 1H-NMR and ultra-performance liquid chromatography-mass spectrometry chemical analyses. Passage of stool from cecum to midcolon was associated with segment-specific changes in fecal AA composition and a decrease in total AA content. Simultaneous measurement of up to 16 AA fluxes under native luminal conditions, with correction for endogenous AA release, demonstrated absorptive transfer of AAs across the cecum and proximal colon at rates comparable (30-80%) to those across the jejunum, with significant Na+-dependent and H+-stimulated components. Expression profiling of 30 major AA transporter genes by quantitative PCR revealed comparatively high levels of transcripts for 20 AA transporters in the cecum and/or colon, with the levels of 12 exceeding those in the small intestine. Our results suggest a more detailed model of major apical and basolateral AA transporters in rat colonocytes and provide evidence for a previously unappreciated transfer of AAs across the colonic epithelium that could link the prodigious metabolic capacities of the luminal microbiota, the colonocytes, and the body tissues.NEW & NOTEWORTHY This study provides evidence for a previously unappreciated transfer of microbially generated amino acids across the colonic epithelium under physiological conditions that could link the prodigious metabolic capacities of the luminal microbiota, the colonocytes, and the body tissues. The segment-specific expression of at least 20 amino acid transporter genes along the colon provides a detailed mechanistic basis for uniport, heteroexchange, Na+-cotransport, and H+-cotransport components of colonic amino acid absorption.
Collapse
Affiliation(s)
- Yuxin Chen
- 1Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Meredith M. Dinges
- 2Department of Chemistry, University of California, Riverside, California
| | - Andrew Green
- 2Department of Chemistry, University of California, Riverside, California
| | - Scott E. Cramer
- 1Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Cynthia K. Larive
- 2Department of Chemistry, University of California, Riverside, California
| | - Christian Lytle
- 1Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| |
Collapse
|
42
|
Amato KR, Jeyakumar T, Poinar H, Gros P. Shifting Climates, Foods, and Diseases: The Human Microbiome through Evolution. Bioessays 2019; 41:e1900034. [PMID: 31524305 DOI: 10.1002/bies.201900034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/29/2019] [Indexed: 12/26/2022]
Abstract
Human evolution has been punctuated by climate anomalies, structuring environments, deadly infections, and altering landscapes. How well humans adapted to these new circumstances had direct effects on fitness and survival. Here, how the gut microbiome could have contributed to human evolutionary success through contributions to host nutritional buffering and infectious disease resistance is reviewed. How changes in human genetics, diet, disease exposure, and social environments almost certainly altered microbial community composition is also explored. Emerging research points to the microbiome as a key player in host responses to environmental change. Therefore, the reciprocal interactions between humans and their microbes are likely to have shaped human patterns of local adaptation throughout our shared evolutionary history. Recent alterations in human lifestyle, however, are altering human microbiomes in unprecedented ways. The consequences of interrupted host-microbe relationships for human adaptive potential in the future are unknown.
Collapse
Affiliation(s)
- Katherine R Amato
- Department of Anthropology, Northwestern University, 1810 Hinman Avenue, Evanston, IL, 60208, USA
| | - Thiviya Jeyakumar
- McGill Center for the Study of Complex Traits, Department of Human Genetics, Department of Biochemistry, McGill University, 3649 Sir William Osler Promenade, Montreal, QC, H3G 0B1, Canada
| | - Hendrik Poinar
- Department of Anthropology, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4M4, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, 3649 Sir William Osler Promenade, Montreal, QC, H3G 0B1, Canada
| |
Collapse
|
43
|
Dietary Fiber and Gut Microbiota in Renal Diets. Nutrients 2019; 11:nu11092149. [PMID: 31505733 PMCID: PMC6770883 DOI: 10.3390/nu11092149] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
Nutrition is crucial for the management of patients affected by chronic kidney disease (CKD) to slow down disease progression and to correct symptoms. The mainstay of the nutritional approach to renal patients is protein restriction coupled with adequate energy supply to prevent malnutrition. However, other aspects of renal diets, including fiber content, can be beneficial. This paper summarizes the latest literature on the role of different types of dietary fiber in CKD, with special attention to gut microbiota and the potential protective role of renal diets. Fibers have been identified based on aqueous solubility, but other features, such as viscosity, fermentability, and bulking effect in the colon should be considered. A proper amount of fiber should be recommended not only in the general population but also in CKD patients, to achieve an adequate composition and metabolism of gut microbiota and to reduce the risks connected with obesity, diabetes, and dyslipidemia.
Collapse
|
44
|
An in vitro analysis of intestinal ammonia transport in fasted and fed freshwater rainbow trout: roles of NKCC, K + channels, and Na +, K + ATPase. J Comp Physiol B 2019; 189:549-566. [PMID: 31486919 DOI: 10.1007/s00360-019-01231-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/15/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022]
Abstract
We examined mechanisms of ammonia handling in the anterior, mid, and posterior intestine of unfed and fed freshwater rainbow trout (Oncorhynchus mykiss), with a focus on the Na+:K+:2Cl- co-transporter (NKCC), Na+:K +-ATPase (NKA), and K+ channels. NKCC was localized by immunohistochemistry to the mucosal (apical) surface of enterocytes, and NKCC mRNA was upregulated after feeding in the anterior and posterior segments. NH4+ was equally potent to K+ in supporting NKA activity in all intestinal sections. In vitro gut sac preparations were employed to examine mucosal ammonia flux rates (Jmamm, disappearance from the mucosal saline), serosal ammonia flux rates (Jsamm, appearance in the serosal saline), and total tissue ammonia production rates (Jtamm = Jsamm - Jmamm). Bumetanide (10-4 mol L-1), a blocker of NKCC, inhibited Jsamm in most preparations, but this was largely due to reduction of Jtamm; Jmamm was significantly inhibited only in the anterior intestine of fed animals. Ouabain (10-4 mol L-1), a blocker of NKA, generally reduced both Jmamm and Jsamm without effects on Jtamm in most preparations, though the anterior intestine was resistant after feeding. Barium (10-2 mol L-1), a blocker of K+ channels, inhibited Jmamm in most preparations, and Jsamm in some, without effects on Jtamm. These pharmacological results, together with responses to manipulations of serosal and mucosal Na+ and K+ concentrations, suggest that NKCC is not as important in ammonia absorption as previously believed. NH4+ appears to be taken up through barium-sensitive K+ channels on the mucosal surface. Mucosal NH4+ uptake via both NKCC and K+ channels is energized by basolateral NKA, which plays an additional role in scavenging NH4+ on the serosal surface to possibly minimize blood toxicity or enhance ion uptake and amino acid synthesis following feeding. Together with recent findings from other studies, we have provided an updated model to describe the current understanding of intestinal ammonia transport in teleost fish.
Collapse
|
45
|
Casati M, Ferri E, Azzolino D, Cesari M, Arosio B. Gut microbiota and physical frailty through the mediation of sarcopenia. Exp Gerontol 2019; 124:110639. [PMID: 31226349 DOI: 10.1016/j.exger.2019.110639] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/18/2022]
Abstract
The changing physiology and lifestyle of older people affect the gut microbiota composition. In particular, the age-related diet modifications can alter the gut microbiota biodiversity and determine the relative abundance of specific microbial taxa, resulting in microbiota dysbiosis with negative consequences for the host physiology. Unhealthy microbiota may then induce an acceleration of the age-related physiological changes, consequently concurring at determining the characteristic complexity of frail older persons. One of the major clinical manifestations of frailty is represented by the individual's physical decline. Besides of a well-established clinical phenotype of frailty, the qualitative and quantitative skeletal muscle impairment (i.e., sarcopenia) is today of particular interest for potentially serving as target for (pharmacological and non-pharmacological) interventions to prevent incident disability. Evidence suggests that gut microbiota is able to influence the skeletal muscle homeostasis via microbiota-dependent metabolites, thus representing the possible biological substratum for the sarcopenia onset. In fact, the rearrangements of gut microbiota as well as the alteration of its functions contribute at increasing the anabolic resistance, releasing pro-inflammatory mediators, determining mitochondrial abnormalities with consequent oxidation, and causing insulin resistance. In this article, the link between gut microbiota and physical frailty is discussed. It is especially explained the role that sarcopenia may play in this likely bidirectional relationship.
Collapse
Affiliation(s)
- Martina Casati
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy.
| | - Evelyn Ferri
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy.
| | - Domenico Azzolino
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| | - Matteo Cesari
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| | - Beatrice Arosio
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| |
Collapse
|
46
|
Tian Z, Ma X, Deng D, Cui Y, Chen W. Influence of Nitrogen Levels on Nutrient Transporters and Regulators of Protein Synthesis in Small Intestinal Enterocytes of Piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:2782-2793. [PMID: 30785738 DOI: 10.1021/acs.jafc.8b06712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To investigate effects of dietary nitrogen level on nutrient absorption and utilization in small intestinal enterocyte of piglets, weaned piglets were fed for 10 days with diets containing 20%, 17%, or 14% crude protein (CP) with supplementation to meet requirements for essential amino acids in vivo, and IPEC-1 cells were cultured with different nitrogen levels (NL) in a culture medium (70%, 85%, and 100%) in vitro by monocultured and cocultured intestinal porcine epithelial cells (IPEC-1) and human gastric epithelial cells (GES-1). The results showed the following: (1) In animal trial, decreased dietary CP reduced transcript abundance of nutrient transporters like CAT1, PepT1, GLUT2, and SGLT-1 in jejunal mucosa (0.09 ± 0.03, P < 0.0001; 0.40 ± 0.04, P = 0.0087; 0.20 ± 0.07, P = 0.0003; 0.35 ± 0.02, P = 0.0001), but 17% CP diet did not affect jejunal protein synthesis. (2) The transcript abundance of nutrient transporters displayed similarly effective tendency in jejunal mucosa and cocultured IPEC-1 rather than that in monocultured IPEC-1. (3) Decreased nitrogen levels reduced expressive abundance of PI3K, Class 3 PI3K, TSC2, and 4E-BP1 in monocultured IPEC-1, but 85% nitrogen level did not affect expressive abundance of PI3K, TSC2, mTORC1, 4E-BP1, and S6K1 in cocultured IPEC-1. In general, decreased 3% CP or 15% nitrogen level reduced relative transcript expression of nutrient transporters, but did not affect protein synthesis in jejunal mucosa and cocultured IPEC-1. Therefore, decreased 3% dietary CP increased utilized and synthetic efficiency of nitrogen resource in small intestine and was beneficial in saving the dietary nitrogen resource.
Collapse
Affiliation(s)
- Zhimei Tian
- Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640 , China
- Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
| | - Xianyong Ma
- Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640 , China
- Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
| | - Dun Deng
- Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640 , China
- Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
| | - Yiyan Cui
- Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640 , China
- Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
| | - Weidong Chen
- Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640 , China
- Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Institute of Animal Science , Guangdong Academy of Agricultural Sciences , Guangzhou 510640 , China
| |
Collapse
|
47
|
Diether NE, Willing BP. Microbial Fermentation of Dietary Protein: An Important Factor in Diet⁻Microbe⁻Host Interaction. Microorganisms 2019; 7:microorganisms7010019. [PMID: 30642098 PMCID: PMC6352118 DOI: 10.3390/microorganisms7010019] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 01/04/2023] Open
Abstract
Protein fermentation by gut microbiota contributes significantly to the metabolite pool in the large intestine and may contribute to host amino acid balance. However, we have a limited understanding of the role that proteolytic metabolites have, both in the gut and in systemic circulation. A review of recent studies paired with findings from previous culture-based experiments suggests an important role for microbial protein fermentation in altering the gut microbiota and generating a diverse range of bioactive molecules which exert wide-ranging host effects. These metabolic products have been shown to increase inflammatory response, tissue permeability, and colitis severity in the gut. They are also implicated in the development of metabolic disease, including obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD). Specific products of proteolytic fermentation such as hydrogen sulfide, ammonia, and p-Cresol may also contribute to the development of colorectal cancer. These findings are in conflict with other studies showing that tryptophan metabolites may improve gut barrier function and attenuate severity in a multiple sclerosis model. Further research examining proteolytic fermentation in the gut may be key to our understanding of how microbial and host metabolism interact affecting health.
Collapse
Affiliation(s)
- Natalie E Diether
- Department of Agriculture, Food & Nutritional Science, University of Alberta, Ag/For Centre, Edmonton Alberta, T6G 2P5, Canada.
| | - Benjamin P Willing
- Department of Agriculture, Food & Nutritional Science, University of Alberta, Ag/For Centre, Edmonton Alberta, T6G 2P5, Canada.
| |
Collapse
|
48
|
Zhou H, Chen D, Mao X, He J, Yu J, Zheng P, Luo J, Gao J, Htoo J, Yu B. Effects of dietary lysine levels on jejunal expression of amino
acids transporters and hindgut microflora in weaned pigs. JOURNAL OF ANIMAL AND FEED SCIENCES 2018. [DOI: 10.22358/jafs/93736/2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
49
|
Wu T, Zhang Y, Lv Y, Li P, Yi D, Wang L, Zhao D, Chen H, Gong J, Hou Y. Beneficial Impact and Molecular Mechanism of Bacillus coagulans on Piglets' Intestine. Int J Mol Sci 2018; 19:ijms19072084. [PMID: 30021943 PMCID: PMC6073773 DOI: 10.3390/ijms19072084] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/27/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023] Open
Abstract
The aim of this research was to investigate the beneficial impact and molecular mechanism of B. coagulans on piglets' intestine. Twenty-four 21 days old weaned piglets were allotted to three treatments: Control group (basal diet), B6 group (basal diet + 2 × 10⁶ CFU/g B. coagulans), and the B7 group (basal diet + 2 × 10⁷ CFU/g B. coagulans). The results showed that, compared with the control group, the B7 group had a reduced cholesterol content and gamma glutamyl transpeptidase (GGT) in plasma (p < 0.05); the B6 and B7 groups had a significantly decreased diarrhea rate and diamine oxidase (DAO) activity in plasma (p < 0.05), increased villus height in ileum and decreased crypt depth in the jejunum (p < 0.05); increased activities of superoxide dismutase (SOD) and catalase (CAT), and decreased the content of malondialdehyde (MDA) and H₂O₂ in the intestine (p < 0.05). These data suggested that supplementing B. coagulans had beneficial impacts on promoting nutrients' metabolism, maintaining intestinal integrity, and alleviating oxidative stress and diarrhea. Further research of molecular mechanisms showed changing expression levels of related proteins and genes, suggesting that these could be involved in the regulation of the impact. The community composition of the gut microbiota was also found to be altered in several operational taxonomic units within the genus, Prevotella (order Bacteroidales), and the order, Clostridiales.
Collapse
Affiliation(s)
- Tao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Yue Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Yang Lv
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Peng Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Dan Yi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Lei Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Di Zhao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Hongbo Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| | - Joshua Gong
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada.
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University (WPHU), Wuhan 430023, China.
| |
Collapse
|
50
|
Levitt DG, Levitt MD. A model of blood-ammonia homeostasis based on a quantitative analysis of nitrogen metabolism in the multiple organs involved in the production, catabolism, and excretion of ammonia in humans. Clin Exp Gastroenterol 2018; 11:193-215. [PMID: 29872332 PMCID: PMC5973424 DOI: 10.2147/ceg.s160921] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Increased blood ammonia (NH3) is an important causative factor in hepatic encephalopathy, and clinical treatment of hepatic encephalopathy is focused on lowering NH3. Ammonia is a central element in intraorgan nitrogen (N) transport, and modeling the factors that determine blood-NH3 concentration is complicated by the need to account for a variety of reactions carried out in multiple organs. This review presents a detailed quantitative analysis of the major factors determining blood-NH3 homeostasis – the N metabolism of urea, NH3, and amino acids by the liver, gastrointestinal system, muscle, kidney, and brain – with the ultimate goal of creating a model that allows for prediction of blood-NH3 concentration. Although enormous amounts of NH3 are produced during normal liver amino-acid metabolism, this NH3 is completely captured by the urea cycle and does not contribute to blood NH3. While some systemic NH3 derives from renal and muscle metabolism, the primary site of blood-NH3 production is the gastrointestinal tract, as evidenced by portal vein-NH3 concentrations that are about three times that of systemic blood. Three mechanisms, in order of quantitative importance, release NH3 in the gut: 1) hydrolysis of urea by bacterial urease, 2) bacterial protein deamination, and 3) intestinal mucosal glutamine metabolism. Although the colon is conventionally assumed to be the major site of gut-NH3 production, evidence is reviewed that indicates that the stomach (via Helicobacter pylori metabolism) and small intestine and may be of greater importance. In healthy subjects, most of this gut NH3 is removed by the liver before reaching the systemic circulation. Using a quantitative model, loss of this “first-pass metabolism” due to portal collateral circulation can account for the hyperammonemia observed in chronic liver disease, and there is usually no need to implicate hepatocyte malfunction. In contrast, in acute hepatic necrosis, hyperammonemia results from damaged hepatocytes. Although muscle-NH3 uptake is normally negligible, it can become important in severe hyperammonemia. The NH3-lowering actions of intestinal antibiotics (rifaximin) and lactulose are discussed in detail, with particular emphasis on the seeming lack of importance of the frequently emphasized acidifying action of lactulose in the colon.
Collapse
Affiliation(s)
- David G Levitt
- Department of Integrative Biology and Physiology, University of Minnesota
| | - Michael D Levitt
- Research Service, Veterans Affairs Medical Center, Minneapolis, MN, USA
| |
Collapse
|