1
|
Voudren CD, Mayhue EJ, Riehm MD, Jugan MC. Evaluation of the relationship between plasma glucagon-like peptide-2 and gastrointestinal dysbiosis in canine chronic enteropathies. PLoS One 2024; 19:e0305711. [PMID: 38935795 PMCID: PMC11210855 DOI: 10.1371/journal.pone.0305711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/04/2024] [Indexed: 06/29/2024] Open
Abstract
Chronic enteropathies are a common cause of morbidity in dogs and are associated with disruption of the normal gastrointestinal mucosal barrier. The objective of this prospective study was to determine the association between measures of gastrointestinal dysbiosis and plasma concentrations of glucagon-like peptide-2, a hormone responsible for normal mucosal structure, in dogs with chronic enteropathies. Fecal 16S V4 rRNA gene sequencing and quantitative PCR via the dysbiosis index was performed on 16 healthy controls and 18 dogs with chronic enteropathy prior to and 1 month after initiation of individualized therapy. Fasting and post-prandial plasma GLP-2 concentrations were measured via ELISA in healthy dogs and chronic enteropathy dogs at both time points. Alpha and beta diversity indices, as well as bacterial population abundances were compared between groups and time-points. Principal component analysis combined with least squares regression was used to identify taxa contributing to glucagon-like peptide-2 variance among groups. While the dysbiosis index did not differ between healthy dogs and dogs with chronic enteropathy, 16S V4 genomic sequencing identified 47 operational taxonomic units that differed between the groups, all but 2 of which resolved following chronic enteropathy treatment. Principal component analysis identified 6 families and 19 genera that contributed to differences in glucagon-like peptide-2 concentrations between groups. Dysbiosis associated with chronic enteropathies in dogs may contribute to the observed lower plasma glucagon-like peptide-2 concentrations. Further research into mechanisms of microbiota impact on the enteroendocrine system is needed. Association between glucagon-like peptide-2 secretion and microbiome indices may help to guide research into future treatment strategies for dogs with chronic enteropathy.
Collapse
Affiliation(s)
- Caylie D. Voudren
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States of America
| | - Erin J. Mayhue
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States of America
| | - Michelle D. Riehm
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States of America
| | - Maria C. Jugan
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States of America
| |
Collapse
|
2
|
Ni M, He H, Chen M, Li Z, Cai H, Chen Z, Li M, Xu H. Supplementation of sodium acetate improves the growth performance and intestinal health of rabbits through Wnt/β-catenin signaling pathway. J Anim Sci 2024; 102:skae197. [PMID: 39037212 PMCID: PMC11337008 DOI: 10.1093/jas/skae197] [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: 03/01/2024] [Accepted: 07/21/2024] [Indexed: 07/23/2024] Open
Abstract
Acetic acid, which is one of the most abundant short-chain fatty acids (SCFA) in rabbits' cecum, has been reported to play an important function during various physiological metabolic processes. The present study was conducted to elucidate the effects of sodium acetate on growth performance and intestinal health by evaluating feed intake and efficiency, diarrhea score, serum and cecum metabolites, cecal pH and SCFA, histological staining, nutritional composition of meat and gene expression profile of cecum in rabbits. As a result of sodium acetate supplement, the feed conversion ratio, diarrhea score, and diameter of muscle fiber were significantly decreased (P < 0.05). Additionally, dietary sodium acetate significantly increased in total area of muscle fibers and content of crude ash (P < 0.05). Dietary sodium acetate significantly increased serum glucose, total bile acid, and total cholesterol levels and decreased amylase, lipase, and tCO2 content (P < 0.05). Further examination suggested that sodium acetate supplementation enhanced the micro-environment of cecum, evidenced by significantly increased levels of total antioxidant capacity, total superoxide dismutase, and glutathione peroxidase, and decreased pH and amylase levels (P < 0.05). According to transcriptome sequencing of cecal tissues, differentially expressed genes were predominantly enriched in cell cycle, ABC transporters, and chemokine signaling pathways. Sodium acetate was further suggested to stimulate the proliferation and migration of rabbits' cecum epithelial cells by activating Wnt/β-catenin pathway both in vivo and in vitro. In conclusion, dietary sodium acetate supplementation improved growth performance and intestinal health in rabbits.
Collapse
Affiliation(s)
- Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
3
|
Mizutani Y, Kawamoto S, Takahashi M, Doi H, Wakida K, Tabuchi S, Tanda M, Soga A, Chijiki R, Takakura H, Kawaguchi K, Higashime A, Watanabe M, Ichikawa H, Matsumoto S, Sakai R, Goto H, Kurata K, Kakiuchi S, Miyata Y, Uryu K, Inui Y, Kitao A, Yakushijin K, Matsuoka H, Minami H. Efficacy and Safety of Synbiotics in Patients Undergoing Autologous Hematopoietic Stem Cell Transplantation: A Randomized, Double-blinded, Placebo-controlled Pilot Study. Intern Med 2023; 62:2949-2958. [PMID: 36792187 PMCID: PMC10641206 DOI: 10.2169/internalmedicine.1314-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/26/2022] [Indexed: 02/16/2023] Open
Abstract
Objective High-dose chemotherapy with autologous hematopoietic stem cell transplantation (auto-HSCT) is an effective treatment option for relapsed and refractory aggressive malignant lymphoma. However, patients frequently experience treatment-induced gastrointestinal symptoms. Synbiotics, including live microorganisms and nondigestible food ingredients, reportedly ameliorate chemotherapy-induced mucosal damage. In this study, we assessed the efficacy and safety of synbiotics in patients undergoing auto-HSCT. Methods This randomized, double-blinded study included patients with malignant lymphoma eligible for auto-HSCT. The patients were randomly assigned to either a synbiotic group receiving Bifidobacterium longum (BB536) and guar gum or a placebo group receiving a placebo containing dextrin. The supplements were administered twice daily from the start of conditioning chemotherapy up to 28 days after auto-HSCT. The primary endpoint was the duration of total parenteral nutrition (TPN). Results In total, 12 patients were included and randomized. The median duration of TPN was 15 (range, 12-33) days in the synbiotic group and 17.5 (range, 0-32) days in the placebo group. The median duration of grade ≥3 diarrhea was shorter in the synbiotic group than in then placebo group (2.5 vs. 6.5 days), as was the duration of hospital stay (31.5 vs. 43 days). The oral intake and quality of life regarding diarrhea and anorexia improved in the synbiotic group after engraftment. Synbiotic infections, including bacteremia, were not observed. Conclusion Synbiotics may reduce gastrointestinal toxicity, thereby reducing nutritional problems and improving the quality of life of patients undergoing auto-HSCT, without severe adverse events.
Collapse
Affiliation(s)
- Yu Mizutani
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Shinichiro Kawamoto
- Department of Transfusion Medicine and Cell Therapy, Kobe University Hospital, Japan
| | | | - Hisayo Doi
- Division of Nursing, Kobe University Hospital, Japan
| | - Kumiko Wakida
- Division of Nutrition, Kobe University Hospital, Japan
| | | | - Masaaki Tanda
- Department of Pharmacy, Kobe University Hospital, Japan
| | - Akihiro Soga
- Department of Pharmacy, Kobe University Hospital, Japan
| | - Ruri Chijiki
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Hidetomo Takakura
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Koji Kawaguchi
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Ako Higashime
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Marika Watanabe
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Hiroya Ichikawa
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Sakuya Matsumoto
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Rina Sakai
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Hideaki Goto
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Keiji Kurata
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Seiji Kakiuchi
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Yoshiharu Miyata
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Kiyoaki Uryu
- Department of Medical Oncology/Hematology, Kobe University Hospital, Japan
| | - Yumiko Inui
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Akihito Kitao
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Kimikazu Yakushijin
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Hiroshi Matsuoka
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| | - Hironobu Minami
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine and Hospital, Japan
| |
Collapse
|
4
|
Ducatelle R, Goossens E, Eeckhaut V, Van Immerseel F. Poultry gut health and beyond. ANIMAL NUTRITION 2023; 13:240-248. [PMID: 37168453 PMCID: PMC10164775 DOI: 10.1016/j.aninu.2023.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023]
Abstract
Intestinal health is critically important for the digestion and absorption of nutrients and thus is a key factor in determining performance. Intestinal health issues are very common in high performing poultry lines due to the high feed intake, which puts pressure on the physiology of the digestive system. Excess nutrients which are not digested and absorbed in the small intestine may trigger dysbiosis, i.e. a shift in the microbiota composition in the intestinal tract. Dysbiosis as well as other stressors elicit an inflammatory response and loss of integrity of the tight junctions between the epithelial cells, leading to gut leakage. In this paper, key factors determining intestinal health and the most important nutritional tools which are available to support intestinal health are reviewed.
Collapse
|
5
|
Bastings JJAJ, Venema K, Blaak EE, Adam TC. Influence of the gut microbiota on satiety signaling. Trends Endocrinol Metab 2023; 34:243-255. [PMID: 36870872 DOI: 10.1016/j.tem.2023.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 03/06/2023]
Abstract
Recent studies show a link between the gut microbiota and the regulation of satiety and energy intake, processes that contribute to the development and pathophysiology of metabolic diseases. However, this link is predominantly established in animal and in vitro studies, whereas human intervention studies are scarce. In this review we focus on recent evidence linking satiety and the gut microbiome, with specific emphasis on gut microbial short-chain fatty acids (SCFAs). Based on a systematic search we provide an overview of human studies linking the intake of prebiotics with gut microbial alterations and satiety signaling. Our outcomes highlight the importance of in-depth examination of the gut microbiota in relation to satiety and provide insights into recent and future studies in this field.
Collapse
Affiliation(s)
- Jacco J A J Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Koen Venema
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands; Centre for Healthy Eating and Food Innovation, Maastricht University, Campus Venlo, Venlo, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Tanja C Adam
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
6
|
Song Y, Wen S, Li F, Fischer-Tlustos A, He Z, Guan LL, Steele M. Metagenomic analysis provides bases on individualized shift of colon microbiome affected by delaying colostrum feeding in neonatal calves. Front Microbiol 2022; 13:1035331. [PMID: 36386713 PMCID: PMC9664197 DOI: 10.3389/fmicb.2022.1035331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
This study investigated the effect of colostrum feeding time on the colon digesta microbiome of 2-day-old dairy calves using whole-genome-based metagenome sequencing, aiming to understand the dynamic changes of the colon microbiome when the colostrum feeding is delayed. In total, 24 male Holstein calves were grouped to different pasteurized colostrum feeding time treatments randomly: TRT0h (45 min after birth, n = 7); TRT6h (6 h after birth, n = 8); and TRT12h (12 h after birth, n = 9). Bacteria, archaea, eukaryotes, and viruses were identified in the colon microbiome, with bacteria (99.20%) being the most predominant domain. Streptococcus, Clostridium, Lactobacillus, Ruminococcus, and Enterococcus were the top five abundant bacteria genera. For colon microbiome functions, 114 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified, with nutrients metabolism-related functions “carbohydrate metabolism,” “amino acid metabolism,” “metabolism of cofactors and vitamins,” “metabolism of terpenoids and polyketides,” and “metabolism of other amino acids” being the top five secondary level of KEGG hierarchy functions. When colon microbiomes were compared, they were not affected by delaying first colostrum feeding at both taxonomic and functional levels. However, distinct clusters of colon microbiome profiles were shown based on PERMANOVA analysis despite of different colostrum feeding treatment, suggesting the individualized responses. Moreover, the relative abundance of microbial taxa, microbial functions, and differentially expressed genes was compared between the two distinct clusters, and different relationships were observed among host differentially expressed genes, differential levels of microbial taxa, and microbial functions between the two clusters. Our results suggest that the host may play an important role in shaping the colon microbiome of neonatal dairy calves in response to the early life feeding management. Whether the observed colon microbiome shifts affect gut health and function in the long term requires further research.
Collapse
Affiliation(s)
- Yang Song
- Animal Nutrition and Feed Science, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- Department of Agriculture, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
| | - Shubo Wen
- Animal Nutrition and Feed Science, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China
| | - Fuyong Li
- Department of Agriculture, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | | | - Zhixiong He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Le Luo Guan
- Department of Agriculture, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Le Luo Guan,
| | - Michael Steele
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
- Michael Steele,
| |
Collapse
|
7
|
Kondo T, Chiba T, Tousen Y. Short-chain fatty acids, acetate and propionate, directly upregulate osteoblastic differentiation. Int J Food Sci Nutr 2022; 73:800-808. [PMID: 35616294 DOI: 10.1080/09637486.2022.2078285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Short-chain fatty acids, including acetate, propionate, and butyrate are metabolites of dietary fibre produced by microbiota in the large intestine, have been proposed to contribute to effects on bone homeostasis. However, it is unclear whether they are used in osteoblasts and directly affect bone formation. We investigated whether short-chain fatty acids are absorbed in osteoblast cells and influence early osteoblastic differentiation using MC3T3-E1 cells. Acetate and propionate upregulated alkaline phosphatase activity, which is an osteoblast differentiation marker, and acetate upregulated alkaline phosphatase mRNA expression after treatment for 9 days, whereas butyrate did not in MC3T3-E1 cells. Butyrate was absorbed more rapidly and to a greater extent than acetate and propionate. These results indicate that short-chain fatty acids were used in osteoblastic cells, and particularly acetate and propionate directly upregulated differentiation in primary osteoblasts. Therefore, acetate and propionate might be useful for maintaining a positive balance of bone turnover.
Collapse
Affiliation(s)
- Takashi Kondo
- Department of Food Function and Labeling, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation and Health and Nutrition, Tokyo, Japan
| | - Tsuyoshi Chiba
- Department of Food Function and Labeling, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation and Health and Nutrition, Tokyo, Japan
| | - Yuko Tousen
- Department of Food Function and Labeling, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation and Health and Nutrition, Tokyo, Japan
| |
Collapse
|
8
|
GOTO T, UMEDA T, HINO S, MORITA T, NISHIMURA N. Oral Intake of Slowly Digestible α-Glucan Such as Resistant Maltodextrin Leads to Increased Secretion of Glucagon-Like Peptide-2 in Rats and Helps Thicken Their Ileal Mucosae. J Nutr Sci Vitaminol (Tokyo) 2022; 68:104-111. [DOI: 10.3177/jnsv.68.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Tomoya GOTO
- Department of Applied Life Sciences, Faculty of Agriculture, Shizuoka University
| | - Tomoki UMEDA
- Graduate School of Integrated Science and Technology, Shizuoka University
| | - Shingo HINO
- College of Agriculture, Academic Institute, Shizuoka University
| | - Tatsuya MORITA
- College of Agriculture, Academic Institute, Shizuoka University
| | | |
Collapse
|
9
|
Fukumori R, Doi K, Mochizuki T, Oikawa S, Gondaira S, Iwasaki T, Izumi K. Sodium butyrate administration modulates the ruminal villus height, inflammation-related gene expression, and plasma hormones concentration in dry cows fed a high-fiber diet. Anim Sci J 2022; 93:e13791. [PMID: 36478496 DOI: 10.1111/asj.13791] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 10/08/2022] [Accepted: 10/24/2022] [Indexed: 12/12/2022]
Abstract
The objectives of this study were to evaluate the effects of sodium butyrate on the ruminal villus morphology, mRNA expression associated with nutrient metabolism and inflammation in the ruminal epithelium, and plasma concentrations of metabolites and hormones in non-lactating cows fed a high-fiber diet. Four Holstein cows with a rumen cannula were assigned to two treatments in a crossover design. The treatments were ruminal administration of sodium butyrate premix or control premix before feeding to cows fed the same total mixed ration mainly composed of glass silage once a day. Sodium butyrate was provided at a butyrate dose of 0.04% per kg body weight. The control premix was made by replacing sodium-butyrate with wheat bran. The plasma β-hydroxybutyrate concentration increased 3 to 6 h after the butyrate premix administration but returned to a concentration similar to that of the control before feeding. After continuous administration, increases in the ruminal villus height and plasma concentration of glucagon-like peptide-2, and lower gene expression of TNF-α, IL-1β, and TLR-2 in the rumen epithelium were observed in cows supplied with the butyrate premix. These results showed that sodium butyrate affects rumen epithelial morphology and plasma concentrations of hormones even under a low fermentable diet.
Collapse
Affiliation(s)
- Rika Fukumori
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Kazuya Doi
- Department of Sustainable Agriculture, College of Agriculture, Food and Environment Sciences, Rakuno Gakuen University, Ebetsu, Japan
| | - Taisei Mochizuki
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Shin Oikawa
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Satoshi Gondaira
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Tomohito Iwasaki
- Department of Food Science and Human Wellness, College of Agriculture, Food and Environment Sciences, Rakuno Gakuen University, Ebetsu, Japan
| | - Kenichi Izumi
- Department of Sustainable Agriculture, College of Agriculture, Food and Environment Sciences, Rakuno Gakuen University, Ebetsu, Japan
| |
Collapse
|
10
|
Samarasinghe MB, Sehested J, Weisbjerg MR, van der Heide ME, Nørgaard JV, Vestergaard M, Hernández-Castellano LE. Feeding milk supplemented with Ulva sp., Ascophyllum nodosum, or Saccharina latissima to preweaning dairy calves: Effects on growth, gut microbiota, gut histomorphology, and short-chain fatty acids in digesta. J Dairy Sci 2021; 104:12117-12126. [PMID: 34454759 DOI: 10.3168/jds.2021-20680] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
Emerging knowledge shows the importance of preweaning nutrition on programming the gastrointestinal microbiome and development of the gut barrier function. The aim of this study was to assess the effects of supplementing cow milk with either intact dried Ulva sp., Ascophyllum nodosum, or Saccharina latissima on growth performance and several gut health parameters of preweaning dairy calves. Forty male Holstein calves were selected based on birth weight (41 ± 4 kg) and plasma Brix percentage (≥8.7%) at d 2 of life. From d 2 to d 42 of life, the control calves (n = 10) were fed with cow milk (8 L/d) without seaweed supplementation, and the experimental calves were fed with cow milk (8 L/d) supplemented with either Ulva sp. (n = 10), A. nodosum (n = 10), or S. latissima (n = 10) at a concentration of 50 g/8 L of cow milk per day (i.e., 5% on a dry matter basis). Calves were weighed every week, and body weight gain and calf starter intake were monitored weekly. At d 42 ± 3 of life, calves were slaughtered. The organ weights and digesta pH from the reticulorumen, mid- and end small intestine, and mid-colon were recorded. A tissue sample (5 cm) collected from the mid-small intestine was analyzed for histomorphology. Digesta from the mid-small intestine and mid-colon were analyzed for lactobacilli, Escherichia coli, and Enterobacteriaceae, and short-chain fatty acid profile. Weight gain of the calves was not affected by seaweed supplementation. Proportional organ weights were not affected by seaweed supplementation except for reticulorumen weight, which was higher in calves fed Ulva sp. Both the mid-small intestinal and mid-colonic digesta populations of lactobacilli, Enterobacteriaceae, and E. coli, as well as the mid-small intestinal histomorphology in seaweed-supplemented calves were not different from control calves. However, acetic acid proportion in mid-colonic digesta was increased in calves fed Ulva sp. and A. nodosum, whereas butyric acid proportion was decreased compared with the control calves. Digesta pH in mid- and end small intestine and mid-colon were not affected, whereas ruminal pH was increased in calves fed Ulva sp. compared with the control calves. In conclusion, intact dried seaweed supplementation did not improve the growth or selected gut health parameters (i.e., histomorphology, digesta pH, bacteria, and short-chain fatty acids) in preweaning Holstein calves.
Collapse
Affiliation(s)
- M B Samarasinghe
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark.
| | - J Sehested
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark
| | - M R Weisbjerg
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark
| | - M E van der Heide
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark
| | - J V Nørgaard
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark
| | - M Vestergaard
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark
| | - L E Hernández-Castellano
- Department of Animal Science, Aarhus University, AU-Foulum, 8830 Tjele, Denmark; Animal Production and Biotechnology group, Institute of Animal Health and Food Safety, Universidad de Las Palmas de Gran Canaria, 35413 Arucas, Spain.
| |
Collapse
|
11
|
Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends Microbiol 2021; 29:700-712. [PMID: 33674141 DOI: 10.1016/j.tim.2021.02.001] [Citation(s) in RCA: 423] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Our ancestral diet consisted of much more nondigestible fiber than that of many societies today. Thus, from an evolutionary perspective the human genome and its physiological and nutritional requirements are not well aligned to modern dietary habits. Fiber reaching the colon is anaerobically fermented by the gut bacteria, which produce short-chain fatty acids (SCFAs) as metabolic by-products. SCFAs play a role in intestinal homeostasis, helping to explain why changes in the microbiota can contribute to the pathophysiology of human diseases. Recent research has shown that SCFAs can also have effects on tissues and organs beyond the gut, through their circulation in the blood. SCFAs not only signal through binding to cognate G-protein-coupled receptors on endocrine and immune cells in the body but also induce epigenetic changes in the genome through effects on the activity of histone acetylase and histone deacetylase enzymes. Furthermore, epigenetic imprinting likely occurs in utero, highlighting the importance of the maternal diet in early life. Here we review current understanding of how SCFAs impact on human and animal physiology and discuss the potential applications of SCFAs in the prevention and treatment of human diseases.
Collapse
|
12
|
Höllwarth ME, Solari V. Nutritional and pharmacological strategy in children with short bowel syndrome. Pediatr Surg Int 2021; 37:1-15. [PMID: 33392698 DOI: 10.1007/s00383-020-04781-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Short bowel syndrome in neonates is a severe and life-threatening disease after a major loss of small bowel with or without large bowel. Intestinal adaptation, by which the organism tries to restore digestive and absorptive capacities, is entirely dependent on stimulation of the active enterocytes by enteral nutrition. This review summarizes recent knowledge about the pathophysiologic consequences after the loss of different intestinal parts and outlines the options for enteral nutrition and pharmacological therapies to support the adaptation process.
Collapse
Affiliation(s)
- Michael E Höllwarth
- Univ. Clinic of Pediatric and Adolescent Surgery, Medical University, Graz, Austria.
| | - Valeria Solari
- Department of Pediatric Surgery, Klinik Donaustadt, 1220, Vienna, Austria
| |
Collapse
|
13
|
FFA2-, but not FFA3-agonists inhibit GSIS of human pseudoislets: a comparative study with mouse islets and rat INS-1E cells. Sci Rep 2020; 10:16497. [PMID: 33020504 PMCID: PMC7536384 DOI: 10.1038/s41598-020-73467-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
The expression of short chain fatty acid receptors FFA2 and FFA3 in pancreatic islets raised interest in using them as drug targets for treating hyperglycemia in humans. This study aims to examine the efficacy of synthetic FFA2- and FFA3-ligands to modulate glucose-stimulated insulin secretion (GSIS) in human pseudoislets which display intact glucose responsiveness. The FFA2-agonists 4-CMTB and TUG-1375 inhibited GSIS, an effect reversed by the FFA2-antagonist CATPB. GSIS itself was not augmented by CATPB. The FFA3-agonists FHQC and 1-MCPC did not affect GSIS in human pseudoislets. For further drug evaluation we used mouse islets. The CATPB-sensitive inhibitory effect of 100 µM 4-CMTB on GSIS was recapitulated. The inhibition was partially sensitive to the Gi/o-protein inhibitor pertussis toxin. A previously described FFA2-dependent increase of GSIS was observed with lower concentrations of 4-CMTB (10 and 30 µM). The stimulatory effect of 4-CMTB on secretion was prevented by the Gq-protein inhibitor FR900359. As in human pseudoislets, in mouse islets relative mRNA levels were FFAR2 > FFAR3 and FFA3-agonists did not affect GSIS. The FFA3-agonists, however, inhibited GSIS in a pertussis toxin-sensitive manner in INS-1E cells and this correlated with relative mRNA levels of Ffar3 > > Ffar2. Thus, in humans, when FFA2-activation impedes GSIS, FFA2-antagonism may reduce glycemia.
Collapse
|
14
|
Fukumori R, Oba M, Izumi K, Otsuka M, Suzuki K, Gondaira S, Higuchi H, Oikawa S. Effects of butyrate supplementation on blood glucagon-like peptide-2 concentration and gastrointestinal functions of lactating dairy cows fed diets differing in starch content. J Dairy Sci 2020; 103:3656-3667. [PMID: 32089297 DOI: 10.3168/jds.2019-17677] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/18/2019] [Indexed: 01/09/2023]
Abstract
The objective of this study was to evaluate effects of butyrate supplementation on plasma concentration of glucagon-like peptide-2 (GLP-2), apparent total-tract digestibility, and responses to a grain challenge of lactating dairy cows fed diets differing in starch content. Eight Holstein cows averaging 58.6 ± 9.96 d in milk (4 primiparous cows fitted with rumen cannula and 4 multiparous intact cows) were blocked by parity and assigned to one of two 4 × 4 Latin squares balanced for carryover effects with a 2 × 2 factorial arrangement of treatments. Treatments were dietary starch content [20.6 vs. 27.5%, respectively, for low starch (LS) and high starch (HS)] and butyrate supplementation (butyrate vs. control) with 21-d periods. Butyrate was provided as Gustor BP70 WS (Norel, S.A., Madrid, Spain), containing 70% sodium butyrate and 30% fatty acid mixture, at 2% of dietary dry matter (providing butyrate at 1.1% of dietary dry matter), and control premix contained 70% wheat bran and 30% fatty acid mixture. Feeds, orts, and fecal samples were collected from d 17 to 19 to determine apparent total-tract nutrient digestibility. Blood and rumen fluid samples were collected on d 19. The baseline of dry matter intake (DMI) was determined as average DMI from d 17 to 19 for each cow, and cows were feed-restricted at 60% of the baseline DMI on d 20, and a grain challenge was conducted by providing steam-flaked corn grain at 0.6% of body weight, on an as-fed basis, in addition to each treatment diet on d 21, and blood and ruminal fluid samples were collected. The interaction of dietary starch content by butyrate supplementation was significant for plasma GLP-2 concentration, being greater for cows fed butyrate with the HS diet than those fed the other 3 diets. Cows fed butyrate increased n-butyrate concentration in the ruminal fluid and tended to increase dry matter and organic matter digestibility compared with the control. During the grain challenge, rumen endotoxin concentration increased over time and was higher for cows fed the HS diets compared with those fed LS diets. However, response variables related to inflammation were not affected by the grain challenge. However, serum haptoglobin, lipopolysaccharide-binding protein, and serum amyloid-A concentrations were greater for cows fed butyrate with the LS diet, but not for those fed the HS diet. These results indicate that butyrate supplementation may increase plasma GLP-2 concentration for cows fed HS diets, and total-tract digestibility regardless of dietary starch content. However, butyrate supplementation did not mitigate inflammation in this study.
Collapse
Affiliation(s)
- R Fukumori
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| | - M Oba
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada T6G 2P5.
| | - K Izumi
- Department of Sustainable Agriculture, College of Agriculture, Food and Environment Sciences, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| | - M Otsuka
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| | - K Suzuki
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| | - S Gondaira
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| | - H Higuchi
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| | - S Oikawa
- Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan 069-8501
| |
Collapse
|
15
|
Yan Y, Xu B, Yin B, Xu X, Niu Y, Tang Y, Wang X, Xie C, Yang T, Zhou S, Yan X, Ma L. Modulation of Gut Microbial Community and Metabolism by Dietary Glycyl-Glutamine Supplementation May Favor Weaning Transition in Piglets. Front Microbiol 2020; 10:3125. [PMID: 32117085 PMCID: PMC7025575 DOI: 10.3389/fmicb.2019.03125] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022] Open
Abstract
Gut microbiota plays a crucial role in diet nutrient metabolism and maintaining host health. The synthetic dipeptides glycyl-glutamine (Gly-Gln) used as diet supplementation to improve the weaning transition of newborns could be metabolized by certain bacteria in vitro. However, the effect of diet Gly-Gln supplementation on gut microbiota in vivo remains largely unknown. 240 piglets at the age of 28 days (day 28) were randomly assigned to two groups that received a basal diet (Ctrl group) or a basal diet supplemented with 0.25% Gly-Gln (Gly-Gln group) for 3 weeks. Five piglets from each group were euthanized for sampling after overnight fasting on day 38 and day 49, respectively. We determined their structure shifts of the gut microbiota using 16S rDNA-based high-throughput sequencing analysis. Microbial metabolites short-chain fatty acids (SCFAs) in the ileum and the colon were determined with high-performance gas chromatography. The concentrations of endocrine peptides including epidermal growth factor, glucagon-like peptide-1, and glucagon-like peptide-2 in ileal mucosa, as well as the serum concentration of interleukin 1 beta, interleukin 6, interleukin 10, and tumor necrosis factor alpha were determined using Enzyme-Linked Immunosorbent Assay. In addition, we also checked the diarrhea ratio, growth performance, and intestinal morphology to assess the favorable effect of dietary Gly-Gln supplementation during the weaning transition. Dietary Gly-Gln supplementation beneficially altered the gut microbiota by increasing bacterial loading, elevating alpha diversity, and increasing the relative abundance of anaerobes and fiber-degrading bacteria (Phylum Fibrobacteres). Accordingly, the microbial metabolites SCFAs in both colon and ileum, as well as the downstream endocrine peptides in the ileum increased. Meanwhile, dietary Gly-Gln's favorable weaning transition was reflected in the increase of growth performance indices and the reduced inflammatory response in a time dependent manner. There were significant correlations among the bacteria which responded to dietary Gly-Gln supplementation and these checked indices. Taken together, dietary Gly-Gln supplementation selectively modulated the gut microbiota, which may favor piglets' weaning-transition. These findings suggest that gut microbiota targeted approaches can be potentially used to improve weaning transition of piglets by dietary functional amino acid.
Collapse
Affiliation(s)
- Yiqin Yan
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Baoyang Xu
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Boqi Yin
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Xiaofan Xu
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Yaorong Niu
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Yimei Tang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Xinkai Wang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Chunlin Xie
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Tao Yang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Shuyi Zhou
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Xianghua Yan
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Libao Ma
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| |
Collapse
|
16
|
Olieman J, Kastelijn W. Nutritional Feeding Strategies in Pediatric Intestinal Failure. Nutrients 2020; 12:E177. [PMID: 31936393 PMCID: PMC7019898 DOI: 10.3390/nu12010177] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/31/2019] [Accepted: 01/04/2020] [Indexed: 12/20/2022] Open
Abstract
Intestinal failure is defined as a critical reduction of the gut mass or function, below the minimum needed to absorb nutrients and fluids. The ultimate goal in intestinal failure is to promote bowel adaptation and reach enteral autonomy while a healthy growth and development is maintained. The condition is heterogeneous and complex. Therefore, recommendations for the type and duration of parenteral, enteral, and oral nutrition are variable, with the child's age as an additional key factor. The aim of this review is to provide an overview of nutritional feeding strategies in this heterogeneous population. Different perspectives on nutritional management, nutrition and adaptation, and microbiome and nutrition will be discussed.
Collapse
Affiliation(s)
- Joanne Olieman
- Department of Internal Medicine, Division of Dietetics, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 40, 3015 GD Rotterdam, The Netherlands;
| | | |
Collapse
|
17
|
Xu B, Yan Y, Huang J, Yin B, Pan Y, Ma L. Cortex Phellodendri extract's anti-diarrhea effect in mice related to its modification of gut microbiota. Biomed Pharmacother 2019; 123:109720. [PMID: 31884345 DOI: 10.1016/j.biopha.2019.109720] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/30/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cortex Phellodendri extract (CPE) has been used in China to treat diarrhea whereas the underlying mechanisms remain poorly understood. Given that dysbiosis of gut microbiota is a potential reason for diarrhea, and that oral CPE has a low absorption rate in intestine, we hypothesized that modification of gut microbiota is an important factor in CPE's anti-diarrhea effect. To test this hypothesis, we established a diarrhea model by challenging post-weaning mice with oral Enterotoxigenic-Escherichia coli (ETEC), and then the mice were treated with two doses of CPE (80 mg/kg bodyweight and 160 mg/kg bodyweight) or the vehicle control (phosphate buffered saline). Diarrhea indices, inflammatory factors, morphology of jejunum, short-chain fatty acids (SCFAs), and serum endocrine were determined. Modification of gut microbiota was analyzed using 16S rDNA high-throughput sequencing. The changes in functional profiles of gut microbiota were predicted using software PICRUSt. We then explored the association between CPE-responding bacteria and the symptoms indices with the spearman's rank correlation coefficient and significance test. Compared with diarrheal mice, CPE decreased Gut/Carcass ratio and water content of stool, increased goblet cell density and villus height/crypt depth of jejunum, as well as decreased inflammatory indices (Tumour Necrosis Factor-α, Myeloperoxidase and Interleukin-1α). CPE shifted the gut microbiota significantly by increasing alpha diversity (observed species, ace, Shannon, and Simpson) and restoring the gut microbiota. CPE increased Firmicutes and decreased Bacteroidetes. The reduced genus Prevotella, Acinetobacter, and Morganella were positively associated with the diarrhea indices, whereas increased genus Odoribacter, Rikenella, and Roseburia were negatively associated with the diarrhea indices. The abundance of carbohydrate metabolism-related gene and SCFAs-producing bacteria were increased, which was evidenced by increased butyric acid and total SCFAs concentration in the caecum. Consequently, endocrine peptides glucagon-like peptide-1, epidermal growth factor, and peptide tyrosine tyrosine in serum were elevated. CONCLUSIONS: CPE shows a shift function on the gut microbiota in alleviating the diarrhea of mice in a dose-dependent manner. In addition, the microbial metabolites SCFAs may mediate CPE's anti-diarrhea effect by enhancing endocrine secretion in mice.
Collapse
Affiliation(s)
- Baoyang Xu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Yiqin Yan
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Juncheng Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Boqi Yin
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Yunxin Pan
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Libao Ma
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China.
| |
Collapse
|
18
|
Yazbeck R, Lindsay RJ, Geier MS, Butler RN, Howarth GS. Prebiotics Fructo-, Galacto-, and Mannan-Oligosaccharide Do Not Protect against 5-Fluorouracil-Induced Intestinal Mucositis in Rats. J Nutr 2019; 149:2164-2173. [PMID: 31504729 DOI: 10.1093/jn/nxz192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/19/2019] [Accepted: 07/24/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Prebiotics selectively stimulate the growth of beneficial bacteria within the gastrointestinal tract, and have been investigated in human and animal studies for their capacity to improve intestinal health. OBJECTIVE We investigated the prebiotics fructo-oligosaccharide (FOS), galacto-oligosaccharide (GOS), and mannan-oligosaccharide (MOS) for their potential to alleviate intestinal damage in rats. METHODS Female Dark Agouti rats (6-8 wk old, 110-150 g) were allocated to 1 of the following treatment groups (n = 8/group): saline/water, saline/FOS, saline/GOS, saline/MOS, 5-fluorouracil (5FU)/water, 5FU/FOS, 5FU/GOS, and 5FU/MOS. Rats were pretreated with either 5% GOS, MOS, or FOS or vehicle (water) from day -12 to day 0. On day 0, rats received a single intraperitoneal injection of saline or 5FU. Metabolic data were recorded daily and all rats were killed on day 3. Histopathology was quantified in hematoxylin and eosin-stained sections. Intestinal sucrase and myeloperoxidase activity were quantified by biochemical assay. Fecal SCFAs-acetic, propionic, and butyric acid-were also measured. Statistical analysis was by repeated-measures, 2-factor ANOVA or Kruskal-Wallis and Mann-Whitney U test; P < 0.05 was considered statistically significant. RESULTS Body weight was significantly decreased in all treatment groups after 5FU injection, with no change in body weight observed in any prebiotic treatment group. Total food intake was lower by ≥7% in the GOS treatment group pre-5FU than in all other groups (P < 0.05). Ileal villus height was 18% higher in GOS-treated rats pre-5FU than in respective water controls (P < 0.05). Jejunal and ileal villus height and crypt depth were significantly decreased in all treatment groups after 5FU injection, with no prebiotic effect observed. SCFAs were differentially increased in prebiotic treatment groups compared with water-only controls (P < 0.05). CONCLUSIONS FOS, GOS, and MOS have differential effects in modifying small intestinal pathology and SCFA profiles in rats with healthy and damaged small intestinal mucosa.
Collapse
Affiliation(s)
- Roger Yazbeck
- College of Medicine and Public Health and Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, South Australia, Australia
| | - Ruth J Lindsay
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, South Australia, Australia
| | - Mark S Geier
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, South Australia, Australia
| | - Ross N Butler
- College of Medicine and Public Health and Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, South Australia, Australia
| | - Gordon S Howarth
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, South Australia, Australia.,Centre for Paediatric and Adolescent Gastroenterology, Children, Youth, and Women's Health Service, North Adelaide, South Australia, Australia
| |
Collapse
|
19
|
Dietary administration of resistant starch improved caecal barrier function by enhancing intestinal morphology and modulating microbiota composition in meat duck. Br J Nutr 2019; 123:172-181. [PMID: 31495347 DOI: 10.1017/s0007114519002319] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Resistant starch (RS) was recently approved to exert a powerful influence on gut health, but the effect of RS on the caecal barrier function in meat ducks has not been well defined. Thus, the effect of raw potato starch (RPS), a widely adopted RS material, on microbial composition and barrier function of caecum for meat ducks was determined. A total of 360 Cherry Valley male ducks of 1-d-old were randomly divided and fed diets with 0 (control), 12, or 24 % RPS for 35 d. Diets supplemented with RPS significantly elevated villus height and villus height:crypt depth ratio in the caecum. The 16S rRNA sequence analysis indicated that the diet with 12 % RPS had a higher relative abundance of Firmicutes and the butyrate-producing bacteria Faecalibacterium, Subdoligranulum, and Erysipelatoclostridium were enriched in all diets. Lactobacillus and Bifidobacterium were significantly increased in the 24 % RPS diet v. the control diet. When compared with the control diet, the diet with 12 % RPS was also found to notably increase acetate, propionate and butyrate contents and up-regulated barrier-related genes including claudin-1, zonula occludens-1, mucin-2 and proglucagon in the caecum. Furthermore, the addition of 12 % RPS significantly reduced plasma TNF-α, IL-1β and endotoxin concentrations. These data revealed that diets supplemented with 12 % RPS partially improved caecal barrier function in meat ducks by enhancing intestinal morphology and barrier markers expression, modulating the microbiota composition and attenuating inflammatory markers.
Collapse
|
20
|
Lambe C, Goulet O, Norsa L. Colon importance in short bowel syndrome. Aging (Albany NY) 2019; 11:9961-9962. [PMID: 31733102 PMCID: PMC6914415 DOI: 10.18632/aging.102447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 10/24/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Cecile Lambe
- Pediatric Gastroenterology Hepatology and Nutrition Necker Enfants Malades Hospital, Paris, France
| | - Olivier Goulet
- Pediatric Gastroenterology Hepatology and Nutrition Necker Enfants Malades Hospital, Paris, France
| | - Lorenzo Norsa
- Pediatric Hepatology Gastroenterology and Transplantation, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
21
|
Kaunitz JD, Akiba Y. Control of Intestinal Epithelial Proliferation and Differentiation: The Microbiome, Enteroendocrine L Cells, Telocytes, Enteric Nerves, and GLP, Too. Dig Dis Sci 2019; 64:2709-2716. [PMID: 31435858 PMCID: PMC7211432 DOI: 10.1007/s10620-019-05778-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jonathan D. Kaunitz
- Medical Service, West Los Angeles VAMC, Los Angeles, CA, USA,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yasutada Akiba
- Research Service, West Los Angeles VAMC, Los Angeles, CA, USA,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
22
|
Karl JP, Armstrong NJ, McClung HL, Player RA, Rood JC, Racicot K, Soares JW, Montain SJ. A diet of U.S. military food rations alters gut microbiota composition and does not increase intestinal permeability. J Nutr Biochem 2019; 72:108217. [PMID: 31473505 DOI: 10.1016/j.jnutbio.2019.108217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022]
Abstract
Interactions between gut microbes and dietary components modulate intestinal permeability (IP) and inflammation. Recent studies have reported altered fecal microbiota composition together with increased IP and inflammation in individuals consuming military food rations in austere environments, but could not isolate effects of the diet from environmental factors. To determine how the U.S. Meal, Ready-to-Eat food ration affects fecal microbiota composition, IP and inflammation, 60 adults (95% male,18-61 years) were randomized to consume their usual ad libitum diet for 31 days (CON) or a strictly controlled Meal, Ready-to-Eat-only diet for 21 days followed by their usual diet for 10 days (MRE). In both groups, fecal microbiota composition was measured before, during (INT, days 1-21) and after the intervention period. IP and inflammation [high-sensitivity C-reactive protein (hsCRP)] were measured on days 0, 10, 21 and 31. Longitudinal changes in fecal microbiota composition differed between groups (P=.005), and fecal samples collected from MRE during INT were identified with 88% accuracy using random forest models. The genera making the strongest contribution to that prediction accuracy included multiple lactic acid bacteria (Lactobacillus, Lactococcus, Leuconostoc), which demonstrated lower relative abundance in MRE, and several genera known to dominate the ileal microbiota (Streptococcus, Veillonella, Clostridium), the latter two demonstrating higher relative abundance in MRE. IP and hsCRP were both lower (34% and 41%, respectively) in MRE relative to CON on day 21 (P<.05) but did not differ otherwise. Findings demonstrate that a Meal, Ready-to-Eat ration diet alters fecal microbiota composition and does not increase IP or inflammation.
Collapse
Affiliation(s)
- J Philip Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, 10 General Greene Ave, Natick, MA 01760, USA.
| | - Nicholes J Armstrong
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, 10 General Greene Ave, Natick, MA 01760, USA.
| | - Holly L McClung
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, 10 General Greene Ave, Natick, MA 01760, USA.
| | - Robert A Player
- Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Rd, Laurel, MD 20723, USA.
| | - Jennifer C Rood
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA.
| | - Kenneth Racicot
- Combat Feeding Directorate, U.S. Army Combat Capabilities Development Command-Soldier Center, 15 General Greene Ave, Natick, MA 01760, USA.
| | - Jason W Soares
- Soldier Performance Optimization Directorate, U.S. Army Combat Capabilities Development Command-Soldier Center, 15 General Greene Ave, Natick, MA 01760, USA.
| | - Scott J Montain
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, 10 General Greene Ave, Natick, MA 01760, USA.
| |
Collapse
|
23
|
Kristek A, Schär MY, Soycan G, Alsharif S, Kuhnle GGC, Walton G, Spencer JPE. The gut microbiota and cardiovascular health benefits: A focus on wholegrain oats. NUTR BULL 2018. [DOI: 10.1111/nbu.12354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- A. Kristek
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - M. Y. Schär
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - G. Soycan
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - S. Alsharif
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - G. G. C. Kuhnle
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - G. Walton
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - J. P. E. Spencer
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| |
Collapse
|
24
|
Herosimczyk A, Lepczyński A, Ożgo M, Barszcz M, Marynowska M, Tuśnio A, Taciak M, Markulen A, Skomiał J. Proteome changes in ileal mucosa of young pigs resulting
from different levels of native chicory inulin in the diet. JOURNAL OF ANIMAL AND FEED SCIENCES 2018. [DOI: 10.22358/jafs/93737/2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
25
|
Neelis E, Koning B, Rings E, Wijnen R, Nichols B, Hulst J, Gerasimidis K. The Gut Microbiome in Patients with Intestinal Failure: Current Evidence and Implications for Clinical Practice. JPEN J Parenter Enteral Nutr 2018; 43:194-205. [DOI: 10.1002/jpen.1423] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Esther Neelis
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Barbara Koning
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Edmond Rings
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
- Paediatric GastroenterologyLeiden University Medical Center–Willem Alexander Children's Hospital Leiden the Netherlands
| | - René Wijnen
- Paediatric SurgeryErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Ben Nichols
- Human NutritionSchool of MedicineCollege of MedicineVeterinary and Life SciencesUniversity of Glasgow Glasgow United Kingdom
| | - Jessie Hulst
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Konstantinos Gerasimidis
- Human NutritionSchool of MedicineCollege of MedicineVeterinary and Life SciencesUniversity of Glasgow Glasgow United Kingdom
| |
Collapse
|
26
|
Ravn J, Glitsø V, Pettersson D, Ducatelle R, Van Immerseel F, Pedersen N. Combined endo -β-1,4-xylanase and α- l -arabinofuranosidase increases butyrate concentration during broiler cecal fermentation of maize glucurono-arabinoxylan. Anim Feed Sci Technol 2018. [DOI: 10.1016/j.anifeedsci.2017.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
27
|
Battson ML, Lee DM, Weir TL, Gentile CL. The gut microbiota as a novel regulator of cardiovascular function and disease. J Nutr Biochem 2017; 56:1-15. [PMID: 29427903 DOI: 10.1016/j.jnutbio.2017.12.010] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
Abstract
The gut microbiome has emerged as a critical regulator of human physiology. Deleterious changes to the composition or number of gut bacteria, commonly referred to as gut dysbiosis, has been linked to the development and progression of numerous diet-related diseases, including cardiovascular disease (CVD). Most CVD risk factors, including aging, obesity, certain dietary patterns, and a sedentary lifestyle, have been shown to induce gut dysbiosis. Dysbiosis is associated with intestinal inflammation and reduced integrity of the gut barrier, which in turn increases circulating levels of bacterial structural components and microbial metabolites that may facilitate the development of CVD. The aim of the current review is to summarize the available data regarding the role of the gut microbiome in regulating CVD function and disease processes. Particular emphasis is placed on nutrition-related alterations in the microbiome, as well as the underlying cellular mechanisms by which the microbiome may alter CVD risk.
Collapse
Affiliation(s)
- Micah L Battson
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO 80523
| | - Dustin M Lee
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO 80523
| | - Tiffany L Weir
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO 80523
| | - Christopher L Gentile
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO 80523.
| |
Collapse
|
28
|
Wang P, Wang Y, Lu L, Yan W, Tao Y, Zhou K, Jia J, Cai W. Alterations in intestinal microbiota relate to intestinal failure-associated liver disease and central line infections. J Pediatr Surg 2017; 52:1318-1326. [PMID: 28501098 DOI: 10.1016/j.jpedsurg.2017.04.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 04/12/2017] [Accepted: 04/29/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The gut microbiota plays a vital role in modulating the metabolic and immune functions of the intestines. We aimed to analyze the dysbiosis of microbiota in infants with short bowel syndrome (SBS) with different complications. PROCEDURE We included 26 fecal samples from 18 infants with SBS during parenteral nutrition. The samples were categorized into three groups: asymptomatic, parenteral nutrition-associated liver disease (PNALD), and central line-associated bloodstream infection (CLABSI). Seven healthy infants were enrolled as controls. Fecal microbiota, secretory IgA, calprotectin, bile acids, and short chain fatty acids were detected. RESULTS The bacterial diversity of the Asymptomatic and Control Groups was significantly higher than that in the PNALD and CLABSI Groups. Proteobacteria was the most pronounced phylum in the PNALD and CLABSI Groups. Decreased acetate was observed in all SBS samples; however, fecal secretory IgA and calprotectin and the proportion of primary and secondary bile acids did not differ from those in healthy controls. CONCLUSIONS Marked alterations of the intestinal microbiota with decreased level of acetate were shown in SBS patients compared with healthy controls. Over-abundance of Proteobacteria (especially Enterobacteriaceae) was found in the samples from the PNALD and CLABSI Groups. LEVEL OF EVIDENCE Prognosis Study, Level I.
Collapse
Affiliation(s)
- Panliang Wang
- Department of Pediatric Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ying Wang
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Lina Lu
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Weihui Yan
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yijing Tao
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Kejun Zhou
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jie Jia
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Department of Nutrition, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei Cai
- Department of Pediatric Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Shanghai Institute of Pediatric Research, Shanghai, China.
| |
Collapse
|
29
|
Elsabagh M, Inabu Y, Obitsu T, Sugino T. Response of plasma glucagon-like peptide-2 to feeding pattern and intraruminal administration of volatile fatty acids in sheep. Domest Anim Endocrinol 2017; 60:31-41. [PMID: 28431319 DOI: 10.1016/j.domaniend.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 03/03/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide-2 (GLP-2), a gut peptide secreted by enteroendocrine L cells, has recently been identified as a key regulator of intestinal growth and absorptive function in ruminants. However, reports on GLP-2 secretion are few, and more information regarding its secretion dynamics is needed. In this study, two experiments were conducted to elucidate the daily rhythm of GLP-2 secretion in response to feeding regimen and to investigate the effect of volatile fatty acids (VFA) on GLP-2 release in sheep. In experiment 1, blood samples were collected over 3 d from 4 Suffolk mature wethers adapted to a maintenance diet fed once daily; day 1 sampling was preceded by 24 h of fasting to reach steady state. On days 1 and 3, samples were collected every 10 min from 11:00 to 14:00 on both days and then every 1 h until 00:00 on day 1 only; feed was offered at 12:00. On day 2, feed was withheld, and sampling was performed every hour from 01:00 to 00:00. In experiment 2, 5 Suffolk mature wethers were assigned to 5 treatment groups of intraruminal administration of saline, acetate, propionate, butyrate, or VFA mix (acetate, propionate, and butyrate in a ratio of 65:20:15) in a 5 × 5 Latin square design. Blood samples were collected at 0, 1.5, 3, 6, 9, 12, 15, 20, 25, 30, 40, 50, 60, 90, and 120 min relative to the beginning of administration at 12:00. In both experiments, plasma GLP-2, glucagon-like peptide-1 (GLP-1), glucose, insulin, and β-hydroxy butyric acid (BHBA) levels were measured. In experiment 1, incremental area under the curve was greater (P < 0.05) post-feeding than pre-feeding on days 1 and 3 for GLP-2 and tended to be greater (P < 0.1) on day 1 for GLP-1. Plasma insulin, glucose, and BHBA levels increased (P < 0.05) on day 1 post-feeding. Plasma GLP-2 was poorly correlated with GLP-1 but positively correlated with insulin, glucose, and BHBA. In experiment 2, administration of butyrate and VFA mix remarkably increased plasma GLP-2 (P = 0.05) and BHBA (P < 0.0001) levels compared with those in other treatments. Plasma GLP-1 levels were higher with butyrate administration compared with those in the saline, acetate, and VFA mix (P = 0.019). Propionate administration increased plasma glucose (P = 0.013) and insulin (P = 0.053) levels. Thus, our data confirmed that GLP-2 release is responsive to feeding and might be promoted by BHBA produced by the rumen epithelial metabolism of butyrate. Further molecular- and cellular-level studies are needed to determine the role of butyrate as a signaling molecule for GLP-2 release.
Collapse
Affiliation(s)
- M Elsabagh
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, 33516 Kafr El-Sheikh, Egypt
| | - Y Inabu
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - T Obitsu
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - T Sugino
- Graduate School of Biosphere Science, The Research Center for Animal Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan.
| |
Collapse
|
30
|
Rodriguez-Castaño GP, Caro-Quintero A, Reyes A, Lizcano F. Advances in Gut Microbiome Research, Opening New Strategies to Cope with a Western Lifestyle. Front Genet 2017; 7:224. [PMID: 28119734 PMCID: PMC5222858 DOI: 10.3389/fgene.2016.00224] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/14/2016] [Indexed: 12/12/2022] Open
Abstract
The "westernization" of global eating and lifestyle habits is associated with the growing rate of chronic diseases, mainly cardiovascular diseases, cancer, type 2 diabetes mellitus, and respiratory diseases. The primary prevention approach is to make nutritional and behavioral changes, however, there is another important determinant of our health that only recently has been considered and is the presence of beneficial microorganisms and their products in our gastrointestinal tract. Microorganisms living in our body can alter the fate of food, drugs, hormones, and xenobiotics, and recent studies point to the use of microorganisms that can counteract the harmful effects of certain compounds introduced or produced endogenously in our body. This review considers the effects of the western lifestyle on adiposity, glucose metabolism, oxidative markers and inflammation profile, emphasizes on the studies that have investigated bacterial strains and products of their metabolism that are beneficial under this lifestyle, and examines the screening strategies that recent studies are using to select the most promising probiotic isolates. In addition, we consider the relevance of studying the microbiota of metabolically healthy people under a western lifestyle for the understanding of the key components that delay the development of chronic diseases.
Collapse
Affiliation(s)
| | - Alejandro Caro-Quintero
- Corporación de Investigación Agropecuaria CORPOICA, Centro de Investigación Tibaitatá Mosquera, Colombia
| | - Alejandro Reyes
- Department of Biological Sciences, Universidad de los AndesBogotá, Colombia; Center for Genome Sciences and Systems Biology, Washington University School of MedicineSt. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of MedicineSt. Louis, MO, USA
| | - Fernando Lizcano
- Center of Biomedical Research, CIBUS, Universidad de La Sabana Chía, Colombia
| |
Collapse
|
31
|
Xu J, Chen HB, Li SL. Understanding the Molecular Mechanisms of the Interplay Between Herbal Medicines and Gut Microbiota. Med Res Rev 2017; 37:1140-1185. [PMID: 28052344 DOI: 10.1002/med.21431] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/21/2016] [Accepted: 11/16/2016] [Indexed: 02/06/2023]
Abstract
Herbal medicines (HMs) are much appreciated for their significant contribution to human survival and reproduction by remedial and prophylactic management of diseases. Defining the scientific basis of HMs will substantiate their value and promote their modernization. Ever-increasing evidence suggests that gut microbiota plays a crucial role in HM therapy by complicated interplay with HM components. This interplay includes such activities as: gut microbiota biotransforming HM chemicals into metabolites that harbor different bioavailability and bioactivity/toxicity from their precursors; HM chemicals improving the composition of gut microbiota, consequently ameliorating its dysfunction as well as associated pathological conditions; and gut microbiota mediating the interactions (synergistic and antagonistic) between the multiple chemicals in HMs. More advanced experimental designs are recommended for future study, such as overall chemical characterization of gut microbiota-metabolized HMs, direct microbial analysis of HM-targeted gut microbiota, and precise gut microbiota research model development. The outcomes of such research can further elucidate the interactions between HMs and gut microbiota, thereby opening a new window for defining the scientific basis of HMs and for guiding HM-based drug discovery.
Collapse
Affiliation(s)
- Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, P.R. China.,Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, 210028, P.R. China
| |
Collapse
|
32
|
Connor EE, Evock-Clover CM, Wall EH, Baldwin RL, Santin-Duran M, Elsasser TH, Bravo DM. Glucagon-like peptide 2 and its beneficial effects on gut function and health in production animals. Domest Anim Endocrinol 2016; 56 Suppl:S56-65. [PMID: 27345324 DOI: 10.1016/j.domaniend.2015.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/17/2015] [Accepted: 11/26/2015] [Indexed: 12/12/2022]
Abstract
Numerous endocrine cell subtypes exist within the intestinal mucosa and produce peptides contributing to the regulation of critical physiological processes including appetite, energy metabolism, gut function, and gut health. The mechanisms of action and the extent of the physiological effects of these enteric peptides are only beginning to be uncovered. One peptide in particular, glucagon-like peptide 2 (GLP-2) produced by enteroendocrine L cells, has been fairly well characterized in rodent and swine models in terms of its ability to improve nutrient absorption and healing of the gut after injury. In fact, a long-acting form of GLP-2 recently has been approved for the management and treatment of human conditions like inflammatory bowel disease and short bowel syndrome. However, novel functions of GLP-2 within the gut continue to be demonstrated, including its beneficial effects on intestinal barrier function and reducing intestinal inflammation. As knowledge continues to grow about GLP-2's effects on the gut and its mechanisms of release, the potential to use GLP-2 to improve gut function and health of food animals becomes increasingly more apparent. Thus, the purpose of this review is to summarize: (1) the current understanding of GLP-2's functions and mechanisms of action within the gut; (2) novel applications of GLP-2 (or stimulators of its release) to improve general health and production performance of food animals; and (3) recent findings, using dairy calves as a model, that suggest the therapeutic potential of GLP-2 to reduce the pathogenesis of intestinal protozoan infections.
Collapse
Affiliation(s)
- E E Connor
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA.
| | - C M Evock-Clover
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - E H Wall
- Pancosma S.A., CH-1218 Geneva, Switzerland
| | - R L Baldwin
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - M Santin-Duran
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - T H Elsasser
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - D M Bravo
- Pancosma S.A., CH-1218 Geneva, Switzerland
| |
Collapse
|
33
|
Akiba Y, Inoue T, Kaji I, Higashiyama M, Narimatsu K, Iwamoto KI, Watanabe M, Guth PH, Engel E, Kuwahara A, Kaunitz JD. Short-chain fatty acid sensing in rat duodenum. J Physiol 2016; 593:585-99. [PMID: 25433076 DOI: 10.1113/jphysiol.2014.280792] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/06/2014] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Luminal lipid in the duodenum modulates gastroduodenal functions via the release of gut hormones and mediators such as cholecystokinin and 5-HT. The effects of luminal short-chain fatty acids (SCFAs) in the foregut are unknown. Free fatty acid receptors (FFARs) for long-chain fatty acids (LCFAs) and SCFAs are expressed in enteroendocrine cells. SCFA receptors, termed FFA2 and FFA3, are expressed in duodenal enterochromaffin cells and L cells, respectively. Activation of LCFA receptor (FFA1) and presumed FFA3 stimulates duodenal HCO3(-) secretion via a glucagon-like peptide (GLP)-2 pathway, whereas FFA2 activation induces HCO3(-) secretion via muscarinic and 5-HT4 receptor activation. The presence of SCFA sensing in the duodenum with GLP-2 and 5-HT signals further supports the hypothesis that luminal SCFA in the foregut may contribute towards the generation of functional symptoms. ABSTRACT Intraduodenal fatty acids (FA) and bacterial overgrowth, which generate short-chain FAs (SCFAs), have been implicated in the generation of functional dyspepsia symptoms. We studied the mechanisms by which luminal SCFA perfusion affects duodenal HCO3(-) secretion (DBS), a measure of mucosal neurohumoral activation. Free fatty acid receptor (FFAR) 1 (FFA1), which binds long-chain FA (LCFA), and SCFA receptors FFA2 and FFA3 were immunolocalised to duodenal enteroendocrine cells. FFA3 colocalised with glucagon-like peptide (GLP)-1, whereas FFA2 colocalised with 5-HT. Luminal perfusion of the SCFA acetate or propionate increased DBS, enhanced by dipeptidyl peptidase-IV (DPPIV) inhibition, at the same time as increasing GLP-2 portal blood concentrations. Acetate-induced DBS was partially inhibited by monocarboxylate/HCO3(-) exchanger inhibition without affecting GLP-2 release, implicating acetate absorption in the partial mediation of DBS. A selective FFA2 agonist dose-dependently increased DBS, unaffected by DPPIV inhibition or by cholecystokinin or 5-HT3 receptor antagonists, but was inhibited by atropine and a 5-HT4 antagonist. By contrast, a selective FFA1 agonist increased DBS accompanied by GLP-2 release, enhanced by DPPIV inhibition and inhibited by a GLP-2 receptor antagonist. Activation of FFA1 by LCFA and presumably FFA3 by SCFA increased DBS via GLP-2 release, whereas FFA2 activation stimulated DBS via muscarinic and 5-HT4 receptor activation. SCFA/HCO3(-) exchange also appears to be present in the duodenum. The presence of duodenal fatty acid sensing receptors that signal hormone release and possibly signal neural activation may be implicated in the pathogenesis of functional dyspepsia.
Collapse
Affiliation(s)
- Yasutada Akiba
- Greater Los Angeles Veterans Affairs Healthcare System, 11301 Wilshire Boulevard, Los Angeles, CA, 90073, USA; Department of Medicine, University of California, Los Angeles, CA, 90095, USA; Brentwood Biomedical Research Institute, 11301 Wilshire Blvd, Los Angeles, CA, 90073, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Onrust L, Ducatelle R, Van Driessche K, De Maesschalck C, Vermeulen K, Haesebrouck F, Eeckhaut V, Van Immerseel F. Steering Endogenous Butyrate Production in the Intestinal Tract of Broilers as a Tool to Improve Gut Health. Front Vet Sci 2015; 2:75. [PMID: 26734618 PMCID: PMC4682374 DOI: 10.3389/fvets.2015.00075] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022] Open
Abstract
The ban on antimicrobial growth promoters and efforts to reduce therapeutic antibiotic usage has led to major problems of gastrointestinal dysbiosis in livestock production in Europe. Control of dysbiosis without the use of antibiotics requires a thorough understanding of the interaction between the microbiota and the host mucosa. The gut microbiota of the healthy chicken is highly diverse, producing various metabolic end products, including gases and fermentation acids. The distal gut knows an abundance of bacteria from within the Firmicutes Clostridium clusters IV and XIVa that produce butyric acid, which is one of the metabolites that are sensed by the host as a signal. The host responds by strengthening the epithelial barrier, reducing inflammation, and increasing the production of mucins and antimicrobial peptides. Stimulating the colonization and growth of butyrate-producing bacteria thus may help optimizing gut health. Various strategies are available to stimulate butyrate production in the distal gut. These include delivery of prebiotic substrates that are broken down by bacteria into smaller molecules which are then used by butyrate producers, a concept called cross-feeding. Xylo-oligosaccharides (XOS) are such compounds as they can be converted to lactate, which is further metabolized to butyrate. Probiotic lactic acid producers can be supplied to support the cross-feeding reactions. Direct feeding of butyrate-producing Clostridium cluster IV and XIVa strains are a future tool provided that large scale production of strictly anaerobic bacteria can be optimized. Current results of strategies that promote butyrate production in the gut are promising. Nevertheless, our current understanding of the intestinal ecosystem is still insufficient, and further research efforts are needed to fully exploit the capacity of these strategies.
Collapse
Affiliation(s)
- Lonneke Onrust
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Richard Ducatelle
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Karolien Van Driessche
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Celine De Maesschalck
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Karen Vermeulen
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Venessa Eeckhaut
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University , Merelbeke , Belgium
| |
Collapse
|
35
|
Sangild PT, Ney DM, Sigalet DL, Vegge A, Burrin D. Animal models of gastrointestinal and liver diseases. Animal models of infant short bowel syndrome: translational relevance and challenges. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1147-68. [PMID: 25342047 PMCID: PMC4269678 DOI: 10.1152/ajpgi.00088.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal failure (IF), due to short bowel syndrome (SBS), results from surgical resection of a major portion of the intestine, leading to reduced nutrient absorption and need for parenteral nutrition (PN). The incidence is highest in infants and relates to preterm birth, necrotizing enterocolitis, atresia, gastroschisis, volvulus, and aganglionosis. Patient outcomes have improved, but there is a need to develop new therapies for SBS and to understand intestinal adaptation after different diseases, resection types, and nutritional and pharmacological interventions. Animal studies are needed to carefully evaluate the cellular mechanisms, safety, and translational relevance of new procedures. Distal intestinal resection, without a functioning colon, results in the most severe complications and adaptation may depend on the age at resection (preterm, term, young, adult). Clinically relevant therapies have recently been suggested from studies in preterm and term PN-dependent SBS piglets, with or without a functional colon. Studies in rats and mice have specifically addressed the fundamental physiological processes underlying adaptation at the cellular level, such as regulation of mucosal proliferation, apoptosis, transport, and digestive enzyme expression, and easily allow exogenous or genetic manipulation of growth factors and their receptors (e.g., glucagon-like peptide 2, growth hormone, insulin-like growth factor 1, epidermal growth factor, keratinocyte growth factor). The greater size of rats, and especially young pigs, is an advantage for testing surgical procedures and nutritional interventions (e.g., PN, milk diets, long-/short-chain lipids, pre- and probiotics). Conversely, newborn pigs (preterm or term) and weanling rats provide better insights into the developmental aspects of treatment for SBS in infants owing to their immature intestines. The review shows that a balance among practical, economical, experimental, and ethical constraints will determine the choice of SBS model for each clinical or basic research question.
Collapse
Affiliation(s)
- Per T. Sangild
- 1Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark; ,2Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark;
| | - Denise M. Ney
- 3Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin;
| | | | - Andreas Vegge
- 1Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark; ,5Diabetes Pharmacology, Novo Nordisk, Måløv, Denmark; and
| | - Douglas Burrin
- 6USDA-ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
36
|
Roy CC, Groleau V, Bouthillier L, Pineault M, Thibault M, Marchand V. Short bowel syndrome in infants: the critical role of luminal nutrients in a management program. Appl Physiol Nutr Metab 2014; 39:745-53. [DOI: 10.1139/apnm-2013-0211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Short bowel syndrome develops when the remnant mass of functioning enterocytes following massive resections cannot support growth or maintain fluid–electrolyte balance and requires parenteral nutrition. Resection itself stimulates the intestine’s inherent ability to adapt morphologically and functionally. The capacity to change is very much related to the high turnover rate of enterocytes and is mediated by several signals; these signals are mediated in large part by enteral nutrition. Early initiation of enteral feeding, close clinical monitoring, and ongoing assessment of intestinal adaptation are key to the prevention of irreversible intestinal failure. The length of the functional small bowel remnant is the most important variable affecting outcome. The major objective of intestinal rehabilitation programs is to achieve early oral nutritional autonomy while maintaining normal growth and nutrition status and minimizing total parenteral nutrition related comorbidities such as chronic progressive liver disease. Remarkable progress has been made in terms of survivability and quality of life, especially in the context of coordinated multidisciplinary programs, but much work remains to be done.
Collapse
Affiliation(s)
- Claude C. Roy
- Département de Pédiatrie, Service de gastroentérologie, hépatologie et nutrition, CHU Sainte-Justine et Université de Montréal, 3175 chemin de la Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Véronique Groleau
- Département de Pédiatrie, Service de gastroentérologie, hépatologie et nutrition, CHU Sainte-Justine et Université de Montréal, 3175 chemin de la Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Lise Bouthillier
- Service de nutrition, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Marjolain Pineault
- Département de pharmacie, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Maxime Thibault
- Département de pharmacie, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Valérie Marchand
- Département de Pédiatrie, Service de gastroentérologie, hépatologie et nutrition, CHU Sainte-Justine et Université de Montréal, 3175 chemin de la Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW It is clear that the metabolic activities of the gut microbiota significantly impact upon human health and disease. RECENT FINDINGS Recent analyses have correlated alterations in microbial community structure with the onset of diabetes, obesity and cardiovascular disease as well as inflammatory conditions of the intestine. This work has demonstrated the influence of diet upon the microbiota in disease states and has identified a number of microbial metabolites that orchestrate the crucial aspects of the host-microbe dialog. The microbial production of short-chain fatty acids, trimethylamine, acetaldehyde and inflammatory mediators has been shown to significantly impact upon the metabolic health of the host through pathways that influence satiety, gut permeability and immune function. In the small intestine, microbial metabolism alters the host bile acid profile affecting the interactions with dedicated bile acid receptors (including FXR and TGR5) to influence both local and systemic cellular responses. Recent findings have, therefore, identified specific microbiota profiles and metabolites as predictors of disease risk as well as determining the microbial species (such as Akkermansia muciniphila and Bilophila wadsworthia) which correlate with health and disease. SUMMARY This work identifies the microbiota as an important target for new diagnostic and therapeutic approaches in metabolic disease.
Collapse
|
38
|
'The way to a man's heart is through his gut microbiota'--dietary pro- and prebiotics for the management of cardiovascular risk. Proc Nutr Soc 2014; 73:172-85. [PMID: 24495527 DOI: 10.1017/s0029665113003911] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human gut microbiota has been identified as a possible novel CVD risk factor. This review aims to summarise recent insights connecting human gut microbiome activities with CVD and how such activities may be modulated by diet. Aberrant gut microbiota profiles have been associated with obesity, type 1 and type 2 diabetes and non-alcoholic fatty liver disease. Transfer of microbiota from obese animals induces metabolic disease and obesity in germ-free animals. Conversely, transfer of pathogen-free microbiota from lean healthy human donors to patients with metabolic disease can increase insulin sensitivity. Not only are aberrant microbiota profiles associated with metabolic disease, but the flux of metabolites derived from gut microbial metabolism of choline, phosphatidylcholine and l-carnitine has been shown to contribute directly to CVD pathology, providing one explanation for increased disease risk of eating too much red meat. Diet, especially high intake of fermentable fibres and plant polyphenols, appears to regulate microbial activities within the gut, supporting regulatory guidelines encouraging increased consumption of whole-plant foods (fruit, vegetables and whole-grain cereals), and providing the scientific rationale for the design of efficacious prebiotics. Similarly, recent human studies with carefully selected probiotic strains show that ingestion of viable microorganisms with the ability to hydrolyse bile salts can lower blood cholesterol, a recognised risk factor in CVD. Taken together such observations raise the intriguing possibility that gut microbiome modulation by whole-plant foods, probiotics and prebiotics may be at the base of healthy eating pyramids advised by regulatory agencies across the globe. In conclusion, dietary strategies which modulate the gut microbiota or their metabolic activities are emerging as efficacious tools for reducing CVD risk and indicate that indeed, the way to a healthy heart may be through a healthy gut microbiota.
Collapse
|
39
|
Castañeda-Gutiérrez E, Moser M, García-Ródenas C, Raymond F, Mansourian R, Rubio-Aliaga I, Viguet-Carrin S, Metairon S, Ammon-Zufferey C, Avanti-Nigro O, Macé K, Silva-Zolezzi I. Effect of a mixture of bovine milk oligosaccharides, Lactobacillus rhamnosus NCC4007 and long-chain polyunsaturated fatty acids on catch-up growth of intra-uterine growth-restricted rats. Acta Physiol (Oxf) 2014; 210:161-73. [PMID: 23834457 DOI: 10.1111/apha.12145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/15/2013] [Accepted: 07/02/2013] [Indexed: 12/21/2022]
Abstract
AIM To investigate the effect of a nutritional mixture (bovine milk oligosaccharides, Lactobacillus rhamnosus NCC4007, arachidonic and docosahexaenoic acid) on growth of intrauterine growth-restricted (IUGR) rats. METHODS IUGR was induced by maternal food restriction. The offspring (males and females) were assigned to: REF (non-IUGR, no mixture), IUGRc (IUGR, no mixture), or IUGRmx (IUGR, mixture). The mixture was given from day 7 to day 58, when tissues and plasma from half of the animals were collected for hormones, metabolites and microarray analysis. The rest received a high-fat diet (HFD) until day 100. Glucose tolerance was measured at 56 and 98 days, and body fat content at 21, 52 and 97 days. RESULTS IUGRmx had the greatest growth during lactation, but from day 22 to day 54, both IUGR groups gained less body weight than the REF (P < 0.05). In the short-term (58 days), IUGRmx tended to be longer (P = 0.06) and had less body fat (P = 0.03) than IUGRc. These differences were not seen after HFD. Microarray analysis of hepatic mRNA expression at 58 and 100 days revealed a gender-dependent treatment effect, and expression of genes related to lipid metabolism was the most affected. Twelve of these genes were selected for studying differences in DNA methylation in the promoter region, for some, we observed age- and gender-related differences but none because of treatment. CONCLUSION The nutritional intervention promoted catch-up growth and normalized excessive adiposity in IUGR animals at short-term. The benefits did not extend after a period of HFD. IUGR and early diet had gender-dependent effects on hepatic gene expression.
Collapse
Affiliation(s)
| | - M. Moser
- Department of Bioanalytical Sciences; Nestlé Research Center; Lausanne Switzerland
| | - C. García-Ródenas
- Department of Nutrition and Health; Nestlé Research Center; Lausanne Switzerland
| | - F. Raymond
- Department of Bioanalytical Sciences; Nestlé Research Center; Lausanne Switzerland
| | - R. Mansourian
- Department of Bioanalytical Sciences; Nestlé Research Center; Lausanne Switzerland
| | - I. Rubio-Aliaga
- Department of Bioanalytical Sciences; Nestlé Research Center; Lausanne Switzerland
| | - S. Viguet-Carrin
- Department of Nutrition and Health; Nestlé Research Center; Lausanne Switzerland
| | - S. Metairon
- Department of Bioanalytical Sciences; Nestlé Research Center; Lausanne Switzerland
| | - C. Ammon-Zufferey
- Department of Nutrition and Health; Nestlé Research Center; Lausanne Switzerland
| | - O. Avanti-Nigro
- Department of Nutrition and Health; Nestlé Research Center; Lausanne Switzerland
| | - K. Macé
- Department of Nutrition and Health; Nestlé Research Center; Lausanne Switzerland
| | - I. Silva-Zolezzi
- Department of Bioanalytical Sciences; Nestlé Research Center; Lausanne Switzerland
| |
Collapse
|
40
|
Brahe LK, Astrup A, Larsen LH. Is butyrate the link between diet, intestinal microbiota and obesity-related metabolic diseases? Obes Rev 2013; 14:950-9. [PMID: 23947604 DOI: 10.1111/obr.12068] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 12/12/2022]
Abstract
It is increasingly recognized that there is a connection between diet, intestinal microbiota, intestinal barrier function and the low-grade inflammation that characterizes the progression from obesity to metabolic disturbances, making dietary strategies to modulate the intestinal environment relevant. In this context, the ability of some Gram-positive anaerobic bacteria to produce the short-chain fatty acid butyrate is interesting. A lower abundance of butyrate-producing bacteria has been associated with metabolic risk in humans, and recent studies suggest that butyrate might have an anti-inflammatory potential that can alleviate obesity-related metabolic complications, possibly due to its ability to enhance the intestinal barrier function. Here, we review and discuss the potential of butyrate as an anti-inflammatory mediator in metabolic diseases, and the potential for dietary interventions increasing the intestinal availability of butyrate.
Collapse
Affiliation(s)
- L K Brahe
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | | | | |
Collapse
|
41
|
Pathobiology and potential therapeutic value of intestinal short-chain fatty acids in gut inflammation and obesity. Dig Dis Sci 2013; 58:2756-66. [PMID: 23839339 PMCID: PMC4317286 DOI: 10.1007/s10620-013-2744-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 06/03/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND The lumen of the gastrointestinal tract contains many substances produced from the breakdown of foodstuffs, from salivary, esophageal, intestinal, hepatic, and pancreatic secretions, and from sloughed cells present in the gastrointestinal lumen. Although these substances were traditionally regarded as waste products, there is increasing realization that many can be biologically active, either as signalling compounds or as nutrients. For example, proteins are broken down into amino acids, which are then sensed by nutrient receptors. The gut microbiome, which is at highest abundance in the ileocecum, has powerful metabolic activity, digesting and breaking down unabsorbed carbohydrates, proteins, and other ingested nutrients into phenols, amines, volatile organic compounds, methane, carbon dioxide, hydrogen, and hydrogen sulfide into volatile fatty acids, also called short-chain fatty acids (SCFAs). CONCLUSION These latter substances are the topic of this review. In this review, we will briefly discuss recent advances in the understanding SCFA production, signalling, and absorption, followed by a detailed description and discussion of trials of SCFAs, probiotics, and prebiotics in the treatment of gastrointestinal disease, in particular ulcerative colitis (UC), pouchitis, short bowel syndrome, and obesity.
Collapse
|
42
|
Fukudome I, Kobayashi M, Dabanaka K, Maeda H, Okamoto K, Okabayashi T, Baba R, Kumagai N, Oba K, Fujita M, Hanazaki K. Diamine oxidase as a marker of intestinal mucosal injury and the effect of soluble dietary fiber on gastrointestinal tract toxicity after intravenous 5-fluorouracil treatment in rats. Med Mol Morphol 2013; 47:100-7. [PMID: 24005798 DOI: 10.1007/s00795-013-0055-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/08/2013] [Indexed: 12/13/2022]
Abstract
The level of plasma diamine oxidase (DAO) activity is associated with the maturation and integrity of small intestinal mucosa. This study in rats investigated whether a decreased level of plasma DAO could reflect the severity of mucosal injury due to intravenous 5-fluorouracil (5-FU) treatment. The beneficial effect of soluble dietary fiber (SDF) on preventing diarrhea after 5-FU treatment was also examined. To induce diarrhea, 5-FU (50 mg/kg/day for four days) was administered via the tail vein with or without SDF supplementation. After 5-FU treatment, the majority of rats developed moderate to severe diarrhea, and levels of plasma DAO activity significantly decreased compared to those of control group (P < 0.05). Scanning electron microscopy revealed disarrangement of the small intestinal villi. Contrarily, the rats supplemented with SDF had diarrhea less frequently (50.0 vs. 91.7 %, P = 0.025) on day five, and DAO activity levels were significantly higher than in those rats administered 5-FU alone (8.25 ± 5.34 vs. 5.50 ± 4.32, P = 0.023). In conclusion, plasma DAO activity decreases in response to severe intestinal mucosal injury after 5-FU treatment, and SDF supplementation might be a practical and useful treatment for reducing the intestinal toxicity of 5-FU.
Collapse
Affiliation(s)
- Ian Fukudome
- Department of Surgery, Kochi Medical School, Kochi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Oozeer R, van Limpt K, Ludwig T, Ben Amor K, Martin R, Wind RD, Boehm G, Knol J. Intestinal microbiology in early life: specific prebiotics can have similar functionalities as human-milk oligosaccharides. Am J Clin Nutr 2013; 98:561S-71S. [PMID: 23824728 DOI: 10.3945/ajcn.112.038893] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human milk is generally accepted as the best nutrition for newborns and has been shown to support the optimal growth and development of infants. On the basis of scientific insights from human-milk research, a specific mixture of nondigestible oligosaccharides has been developed, with the aim to improve the intestinal microbiota in early life. The mixture has been extensively studied and has been shown to be safe and to have potential health benefits that are similar to those of human milk. The specific mixture of short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides has been found to affect the development of early microbiota and to increase the Bifidobacterium amounts as observed in human-milk-fed infants. The resulting gut ecophysiology is characterized by high concentrations of lactate, a slightly acidic pH, and specific short-chain fatty acid profiles, which are high in acetate and low in butyrate and propionate. Here, we have summarized the main findings of dietary interventions with these specific oligosaccharides on the gut microbiota in early life. The gut ecophysiology in early life may have consequences for the metabolic, immunologic, and even neurologic development of the child because reports increasingly substantiate the important function of gut microbes in human health. This review highlights major findings in the field of early gut colonization and the potential impact of early nutrition in healthy growth and development.
Collapse
Affiliation(s)
- Raish Oozeer
- Danone Research-Centre for Specialised Nutrition, Wageningen, Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Effect of supplementary calcium phosphate on plasma gastrointestinal hormones in a double-blind, placebo-controlled, cross-over human study. Br J Nutr 2013; 111:287-93. [DOI: 10.1017/s0007114513002341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Gastrointestinal hormones and Ca are associated with bone metabolism. The objective of the present human study was to determine the effect of calcium phosphate on the postprandial circulation of gastrointestinal hormones. A total of ten men participated in the present double-blind, placebo-controlled, cross-over study. The participants were divided into two groups. Of these, one group consumed bread enriched with 1 g Ca (pentacalcium hydroxy-triphosphate, CaP) daily for 3 weeks. The other group consumed placebo bread. After 2 weeks of washout, the intervention was changed between the groups for another 3 weeks. The subjects consumed a defined diet at the beginning (single administration) and at the end (repeated administration) of the intervention periods, and blood samples were drawn at 0, 30, 60, 120, 180 and 240 min. Between 0 and 30 min, the participants consumed a test meal, with or without CaP. The concentrations of gastrointestinal hormones (glucose-dependent insulinotropic polypeptide, glucagon-like peptide (GLP) 1 and GLP2), insulin and glucose were determined. The AUC of GLP1 (total and active) and GLP2 increased significantly after the repeated CaP administrations compared with that after placebo administration. The AUC of insulin and glucose showed no differences between the CaP and placebo administrations. CaP affects the postprandial plasma concentrations of gastrointestinal hormones through the modulation of the intestinal environment, e.g. bile acids and microbiota.
Collapse
|
45
|
Hua Z, Turner JM, Mager DR, Sigalet DL, Wizzard PR, Nation PN, Ball RO, Pencharz PB, Wales PW. Effects of polymeric formula vs elemental formula in neonatal piglets with short bowel syndrome. JPEN J Parenter Enteral Nutr 2013; 38:498-506. [PMID: 23690158 DOI: 10.1177/0148607113489151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Intestinal adaptation is important for recovery in short bowel syndrome (SBS). This process is dependent on the presence of enteral nutrition (EN) and trophic factors, such as glucagon-like peptide-2 (GLP-2). In clinical practice, elemental formula is often used to feed neonates with SBS, whereas animal studies suggest polymeric formula promotes better intestinal adaptation. In neonatal piglet models of SBS, with or without ileum, we compared the elemental with the polymeric formula, including the effect on endogenous GLP-2. MATERIALS AND METHODS Forty-eight piglets underwent 75% mid-intestinal resection with jejunoileal anastomosis, 75% distal-intestinal resection with jejunocolic anastomosis (JC), or sham without resection. Parenteral nutrition (PN) started postoperatively, tapering as EN was increased, according to clinical criteria, based on diarrhea and weight. Within groups, piglets were randomized to an isocaloric/isonitrogenous elemental (amino acid) or polymeric (intact protein) diet. Plasma GLP-2 and histology for adaptation were measured at 14 days. RESULTS Within both SBS and control groups, no difference in adaptation was observed according to diet. A difference was observed only within the JC piglet group with regard to clinical outcomes. In these piglets, compared with elemental formula, the polymeric formula was associated with more diarrhea ( P = .023) and longer duration of PN support (P = .047). CONCLUSION An overall benefit of the polymeric formula over the elemental formula on gut adaptation was not observed. Furthermore, SBS piglets without ileum had less ability to tolerate polymeric formula, contributing to more days of PN support.
Collapse
Affiliation(s)
- Zheng Hua
- Department of Paediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sato S, Hokari R, Kurihara C, Sato H, Narimatsu K, Hozumi H, Ueda T, Higashiyama M, Okada Y, Watanabe C, Komoto S, Tomita K, Kawaguchi A, Nagao S, Miura S. Dietary lipids and sweeteners regulate glucagon-like peptide-2 secretion. Am J Physiol Gastrointest Liver Physiol 2013; 304:G708-14. [PMID: 23370677 DOI: 10.1152/ajpgi.00282.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucagon-like peptide-2 (GLP-2) is a potent intestinal growth factor derived from enteroendocrine L cells. Although food intake is known to increase GLP-2 secretion, its regulatory mechanisms are largely unknown as a result of its very short half-life in venules. The aims of this study were to compare the effects of luminal nutrients on the stimulation of GLP-2 secretion in vivo using lymph samples and to clarify the involvement of the sweet taste receptor in this process in vitro. Lymph samples were collected from the thoracic duct after bolus administration of dietary lipids or sweetening agents into the duodenum of rats. Human enteroendocrine NCI-H716 cells were also used to compare the effects of various nutrients on GLP-2 secretion. GLP-2 concentrations were measured by ELISA in vivo and in vitro. GLP-2 secretion was enhanced by polyunsaturated fatty acid- and monounsaturated fatty acid-rich dietary oils, dietary carbohydrates, and some kinds of sweeteners in rats; this effect was reproduced in NCI-H716 cells using α-linolenic acid (αLA), glucose, and sweeteners. GLP-2 secretion induced by sweetening agents was inhibited by lactisole, a sweetness-antagonizing inhibitor of T1R3. In contrast, lactisole was unable to inhibit GLP-2 secretion induced by αLA alone. Our results suggested that fatty acid- and sweetener-induced GLP-2 secretion may be mediated by two different pathways, with the sweet taste receptor involved in the regulation of the latter.
Collapse
Affiliation(s)
- Shingo Sato
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Dietary nutrients are essential for gastrointestinal (GI) growth and function, and nutritional support of GI growth and development is a significant component of infant care. For healthy full-term neonates, nutritional provisions of the mother's milk and/or formula will support normal maturation of structure and function of the GI tract in most infants. The composition of breast milk affects GI barrier function and development of a competent mucosal immune system. The functional nutrients and other bioactive components of milk support a microenvironment for gut protection and maturation. However, premature infants struggle with feeding tolerance impairing normal GI function, leading to intestinal dysfunction and even death. The high prevalence worldwide of enteric diseases and dysfunction in neonates has led to much interest in understanding the role of nutrients and food components in the establishment and maintenance of a functioning GI tract. Neonates who do not receive enteral feeding as either mother's milk or formula are supported by total parental nutrition (TPN). The lack of enteral nutrition can compound intestinal dysfunction, leading to high morbidity and mortality in intestinally compromised infants. Reciprocally, enteral stimulation of an immature GI tract can also compound intestinal dysfunction. Therefore, further understanding of nutrient interactions with the mucosa is necessary to define nutritional requirements of the developing GI tract to minimize intestinal complications and infant morbidity. Piglet models of intestinal development and function are similar to humans, and this review summarizes recent findings regarding nutrient requirements for growth and maintenance of intestinal health. In particular, this article reviews the role of specific amino acids (arginine, glutamine, glutamate, and threonine), fatty acids (long chain polyunsaturated, medium chain, and short chain), various prebiotic carbohydrates (short-chain fructo-oligosaccharide, fructo--oligosaccharide, lacto-N-neotetraose, human milk oligosaccharide, polydextrose, and galacto-oligosaccharide), and probiotics that have been examined in the suckling piglet model of intestinal health.
Collapse
Affiliation(s)
- Sheila K Jacobi
- Laboratory of Developmental Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
48
|
Yamamoto Y, Hiasa Y, Murakami H, Ikeda Y, Yamanishi H, Abe M, Matsuura B, Onji M. Rapid alternative absorption of dietary long-chain fatty acids with upregulation of intestinal glycosylated CD36 in liver cirrhosis. Am J Clin Nutr 2012; 96:90-101. [PMID: 22648712 DOI: 10.3945/ajcn.111.033084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Dietary long-chain fatty acid (LCFA) intake is an important risk factor for hepatic inflammation and hepatocarcinogenesis. An alternate route of dietary LCFA absorption has been suggested in patients with liver cirrhosis (LC). OBJECTIVE We aimed to determine this alternate route and to identify its mechanism. DESIGN Twenty healthy control subjects and 47 patients with LC-n = 23 with portal hypertension [PH(+)LC] and 24 without portal hypertension [PH(-)LC)]-were enrolled. [¹³C]Palmitate (an LCFA) and octanoate (a medium-chain fatty acid [MCFA]) were administered by using gastrointestinal endoscopy. Breath ¹³CO₂ was measured to quantify metabolized fatty acids. We also examined intestinal specimens of patients in these groups. RESULTS A more rapid increase in metabolized palmitate, which showed a pattern similar to that of octanoate metabolism, was observed in patients with LC than in healthy control subjects. The increase in the PH(-)LC group was higher than that in the PH(+)LC group. However, the concentration of metabolized palmitate increased with treatment of the PH(+)LC group with a portal-systemic shunt. Morphologic changes such as expanded lymph and blood vessels were present, and glycosylated CD36 increased in the jejunum of the PH(+)LC group. This group had high serum concentrations of glucagon-like peptide-2. These data suggest that dietary LCFAs, similar to MCFAs, are absorbed via blood vessels in patients with LC. CONCLUSIONS Rapid absorption of LCFAs by an alternative method occurred in patients with LC. This altered LCFA processing is likely related to upregulation of intestinal glycosylated CD36 and could contribute to pathogenesis in patients with LC.
Collapse
Affiliation(s)
- Yasunori Yamamoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Current status of pediatric intestinal failure, rehabilitation, and transplantation: summary of a colloquium. Transplantation 2012; 92:1173-80. [PMID: 22067308 DOI: 10.1097/tp.0b013e318234c325] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
An international symposium convened September 9-11, 2010, in Chicago to present the state of the art and science of the multidisciplinary care of intestinal failure in children. Medical and surgical management of the child with intestinal failure was presented with a focus on the importance of multidisciplinary intestinal failure management. Issues of timing of referral and benefit risk analysis for intestine "rehabilitation" and transplant were presented. Areas of opportunity such as increased donor recovery, improvement of long-term transplant outcomes, optimization of immune monitoring, and quality-of-life outcomes were reviewed.
Collapse
|
50
|
Cannon S, Fahey G, Pope L, Bauer L, Wallace R, Miller B, Drackley J. Inclusion of psyllium in milk replacer for neonatal calves. 2. Effects on volatile fatty acid concentrations, microbial populations, and gastrointestinal tract size. J Dairy Sci 2010; 93:4744-58. [DOI: 10.3168/jds.2010-3077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 05/17/2010] [Indexed: 11/19/2022]
|