51
|
Butera A, Di Paola M, Vitali F, De Nitto D, Covotta F, Borrini F, Pica R, De Filippo C, Cavalieri D, Giuliani A, Pronio A, Boirivant M. IL-13 mRNA Tissue Content Identifies Two Subsets of Adult Ulcerative Colitis Patients With Different Clinical and Mucosa-Associated Microbiota Profiles. J Crohns Colitis 2020; 14:369-380. [PMID: 31501882 DOI: 10.1093/ecco-jcc/jjz154] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS A personalized approach to therapy hold great promise to improve disease outcomes. To this end, the identification of different subsets of patients according to the prevalent pathogenic process might guide the choice of therapeutic strategy. We hypothesize that ulcerative colitis [UC] patients might be stratified according to distinctive cytokine profiles and/or to a specific mucosa-associated microbiota. METHODS In a cohort of clinically and endoscopic active UC patients and controls, we used quantitative PCR to analyse the mucosal cytokine mRNA content and 16S rRNA gene sequencing to assess the mucosa-associated microbiota composition. RESULTS We demonstrate, by means of data-driven approach, the existence of a specific UC patient subgroup characterized by elevated IL-13 mRNA tissue content separate from patients with low IL-13 mRNA tissue content. The two subsets differ in clinical-pathological characteristics. High IL-13 mRNA patients are younger at diagnosis and have a higher prevalence of extensive colitis than low IL-13 mRNA patients. They also show more frequent use of steroid/immunosuppressant/anti-tumour necrosis factor α therapy during 1 year of follow-up. The two subgroups show differential enrichment of mucosa-associated microbiota genera with a prevalence of Prevotella in patients with high IL-13 mRNA tissue content and Sutterella and Acidaminococcus in patients with low IL-13 mRNA tissue content. CONCLUSION Assessment of mucosal IL-13 mRNA might help in the identification of a patient subgroup that might benefit from a therapeutic approach modulating IL-13. PODCAST This article has an associated podcast which can be accessed at https://academic.oup.com/ecco-jcc/pages/podcast.
Collapse
Affiliation(s)
- Alessia Butera
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Rome, Italy
| | - Monica Di Paola
- Department of Biology, University of Florence, Firenze, Italy
| | - Francesco Vitali
- Institute of Agricultural Biology and Biotechnology, National Research Council, Pisa, Italy
| | | | - Francesco Covotta
- University "Sapienza", Dept General Surgery, "P. Stefanini", Rome, Italy
| | | | - Roberta Pica
- Sandro Pertini Hospital, IBD, GE Unit, Rome, Italy
| | - Carlotta De Filippo
- Institute of Agricultural Biology and Biotechnology, National Research Council, Pisa, Italy
| | | | | | - Annamaria Pronio
- University "Sapienza", Dept General Surgery, "P. Stefanini", Rome, Italy
| | - Monica Boirivant
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Rome, Italy
| |
Collapse
|
52
|
Far infrared radiation induces changes in gut microbiota and activates GPCRs in mice. J Adv Res 2020; 22:145-152. [PMID: 31969995 PMCID: PMC6965508 DOI: 10.1016/j.jare.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/28/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022] Open
Abstract
Transient exposure of FIR induced compositional and temporal changes in gut microbiota of mice. FIR exposure stimulated growth of the gut beneficial bacteria. FIR exposure promoted growth of the gut SCFAs-producing bacteria. FIR treatment upregulated the expressions of the SCFAs-sensing G-protein coupled receptors in the intestinal mucous of the mice.
Far infrared radiation (FIR) has been widely used to treat chronic diseases and symptoms; however, the underlying mechanism remains unclear. As gut microbiota (GM) markedly impact the host’s physiology, making GM a potential target for the therapeutic evaluation of FIR. C57BL/6J mice were exposed to five times of 2 min-FIR exposure on the abdomen, with a two-hour interval of each exposure within one day. Fecal samples were collected on day one and day 25 after the FIR/control treatment, and the extracted fecal DNAs were evaluated using ERIC-PCR and 16S amplicon sequencing. Host’s G-protein coupled receptors (GPCR) were analyzed using qRT-PCR. FIR induced immediate changes in the GM composition. A prompt and significant (p < 0.05) reduction in the abundance of phylum Deferribacteres (comprised of several pathogens) was observed in the FIR-irradiated mice compared to the control group. Contrarily, FIR exposure induced beneficial genera such as Alistipes, Barnesiella, and Prevotella. The gut of FIR-irradiated mice was predominated by short-chain fatty acids (SCFAs) producers. Also, FIR stimulated the expression of SCFAs-sensing receptors, GPCR 41, 43, and 109 in the gut epithelial barrier. These findings provide the first-hand evidence in which the beneficial effects of FIR radiation might be partially through the modulation of GM.
Collapse
|
53
|
van Niekerk JK, Fischer-Tlustos AJ, Deikun LL, Quigley JD, Dennis TS, Suarez-Mena FX, Hill TM, Schlotterbeck RL, Guan LL, Steele MA. Effect of amount of milk replacer fed and the processing of corn in starter on growth performance, nutrient digestibility, and rumen and fecal fibrolytic bacteria of dairy calves. J Dairy Sci 2020; 103:2186-2199. [PMID: 31954563 DOI: 10.3168/jds.2019-17372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/14/2019] [Indexed: 11/19/2022]
Abstract
The aim of this study was to evaluate effects of milk replacer (MR) feeding rate and processing of corn in calf starter (CS) on growth performance, nutrient digestibility, and rumen and fecal fibrolytic bacteria in dairy calves. Holstein male calves (n = 48, 2-3 d of age) were randomly assigned to 1 of 4 treatments with a 2 × 2 factorial arrangement of MR level of 0.749 kg of MR/d (LO) or up to 1.498 kg of MR/d (HI); and whole corn or flaked corn in textured CS. Calves were weaned by reducing MR offered by 50% during wk 6. Intakes of MR and CS were recorded daily, whereas body weight (BW) was measured weekly. Rumen fluid and fecal matter were collected at wk 5 and 8 to quantify fibrolytic bacteria and nutrient digestibility. Data were analyzed as a completely randomized design using mixed model ANOVA. Repeated measures were used as appropriate. Calves fed HI had greater average daily gain than calves fed LO at wk 2, 3, 4, and 5, yet at wk 7 calves fed HI had lower average daily gain compared with calves fed LO. Starter intake was greater for calves fed LO compared with HI at wk 4, 5, 6, and 7. During wk 5 and 8, calves fed LO had increased ADF and NDF digestibility compared with calves fed HI. During wk 5, dry matter and organic matter digestibility were lower for LO-fed calves compared with HI-fed calves, but during wk 8 the opposite was observed, with HI-fed calves having lower dry matter and organic matter digestibility than LO-fed calves. At wk 5, Clostridium cluster IV and Butyrivibrio fibrisolvens proportions in rumen fluid tended to be higher and Clostridium cluster IV, Fecalibacterium sp., and Prevotella sp. proportions in fecal matter were higher in calves fed LO compared with HI. From wk 8 to 16, dry matter intake was unaffected by treatment; however, energy efficiency was greater in calves fed LO, causing LO calves to have higher BW gain during this period. Greater starter digestibility was observed for calves fed LO versus HI in concert with increased fibrolytic bacteria proportions (wk 5) in fecal and rumen samples, which resulted in greater postweaning BW gain and similar BW and frame measurements by 16 wk of age. Overall the results show that rate of MR feeding has a larger effect than the processing of corn in CS on performance, fiber digestibility, and rumen and fecal fibrolytic bacterial communities.
Collapse
Affiliation(s)
- J K van Niekerk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - A J Fischer-Tlustos
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - L L Deikun
- Nurture Research Center, Provimi, Cargill Animal Nutrition, Brookville, OH 45309
| | - J D Quigley
- Nurture Research Center, Provimi, Cargill Animal Nutrition, Brookville, OH 45309
| | - T S Dennis
- Nurture Research Center, Provimi, Cargill Animal Nutrition, Brookville, OH 45309
| | - F X Suarez-Mena
- Nurture Research Center, Provimi, Cargill Animal Nutrition, Brookville, OH 45309
| | - T M Hill
- Nurture Research Center, Provimi, Cargill Animal Nutrition, Brookville, OH 45309
| | - R L Schlotterbeck
- Nurture Research Center, Provimi, Cargill Animal Nutrition, Brookville, OH 45309
| | - L L Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - M A Steele
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada.
| |
Collapse
|
54
|
Nyman M, Nguyen TD, Wikman O, Hjortswang H, Hallert C. Oat Bran Increased Fecal Butyrate and Prevented Gastrointestinal Symptoms in Patients With Quiescent Ulcerative Colitis-Randomized Controlled Trial. CROHN'S & COLITIS 360 2020; 2:otaa005. [PMID: 36777965 PMCID: PMC9802401 DOI: 10.1093/crocol/otaa005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Indexed: 12/23/2022] Open
Abstract
Background Oat bran specifically increases colon butyrate concentrations and could therefore affect the progress of the disease in patients with ulcerative colitis (UC). Methods Patients with UC in remission were enrolled in a controlled multicenter study and randomized to eat oat bran or low-fiber wheat products. Results Ninety-four of the enrolled patients (n = 47 for both groups) completed the 24-week study. The oat bran group had significantly (P < 0.05) higher fecal butyrate concentrations and lower serum LDL levels, while deterioration of gastrointestinal symptoms was prevented, and subjective health maintained. The control diet significantly (P < 0.05) increased obstipation, reflux, and the symptom burden and had no effects on butyrate or LDL-cholesterol. The relapse rate was the same for both diets. Conclusions Oat bran was well tolerated when given to patients with quiescent UC.
Collapse
Affiliation(s)
- Margareta Nyman
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Thao Duy Nguyen
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | | | - Henrik Hjortswang
- Department of Gastroenterology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Claes Hallert
- Gastroenterology Unit at Norrköping Hospital, Norrköping, Sweden
| |
Collapse
|
55
|
Abstract
The last decade has been characterized by an intense research on the composition of the gut microbiome and the links with human health. While previous work was focused on the effects of prebiotics and probiotics, nowadays several laboratories are describing the gut microbiome and its metabolic functions. Gut microbiome interaction with nutrients allows the gut microbiome to survive and at the same time determines the production of metabolites that are either adsorbed by intestinal cell in a mutual relationship or promote detrimental effect. Metabolomics, a new method to approach identification of biomarkers has been used to identify small metabolites in blood and other biofluids. The study of metabolome revealed several microbial derived metabolites that are circulating in blood and potentially affect human health. In this review we describe the links between regulation of metabolism and microbial derived metabolites.
Collapse
Affiliation(s)
- Ben Arpad Kappel
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
56
|
Shinohara R, Sasaki K, Inoue J, Hoshi N, Fukuda I, Sasaki D, Kondo A, Osawa R. Butyryl-CoA:acetate CoA-transferase gene associated with the genus Roseburia is decreased in the gut microbiota of Japanese patients with ulcerative colitis. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2019; 38:159-163. [PMID: 31763119 PMCID: PMC6856515 DOI: 10.12938/bmfh.18-029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/10/2019] [Indexed: 11/05/2022]
Abstract
Microbial production of butyrate is impaired in patients with ulcerative colitis (UC); however, this inhibition is not well understood in Japanese UC patients. Therefore, we quantitatively analyzed genes encoding butyryl-CoA:acetate CoA-transferase (but) and butyrate kinase (buk) in the gut microbiota of Japanese patients with UC and healthy volunteers (HVs). But showed higher levels than buk. Moreover, patients with UC showed significantly decreased levels of but associated with Roseburia sp./Eubacterium rectale compared with HVs. But, which is associated with Faecalibacterium sp., was maintained in patients with UC, with an unchanged relative abundance of Faecalibacterium sp. microorganisms in patients with UC compared with HVs.
Collapse
Affiliation(s)
- Ryohei Shinohara
- Division Food and Drug Evaluation Science, Department of Social/Community Medicine and Health Science, Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Kengo Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| | - Jun Inoue
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Itsuko Fukuda
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan.,Research Center for Food Safety and Security, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| | - Daisuke Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan.,RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Ro Osawa
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan.,Research Center for Food Safety and Security, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| |
Collapse
|
57
|
Kaplan RC, Wang Z, Usyk M, Sotres-Alvarez D, Daviglus ML, Schneiderman N, Talavera GA, Gellman MD, Thyagarajan B, Moon JY, Vázquez-Baeza Y, McDonald D, Williams-Nguyen JS, Wu MC, North KE, Shaffer J, Sollecito CC, Qi Q, Isasi CR, Wang T, Knight R, Burk RD. Gut microbiome composition in the Hispanic Community Health Study/Study of Latinos is shaped by geographic relocation, environmental factors, and obesity. Genome Biol 2019; 20:219. [PMID: 31672155 PMCID: PMC6824043 DOI: 10.1186/s13059-019-1831-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hispanics living in the USA may have unrecognized potential birthplace and lifestyle influences on the gut microbiome. We report a cross-sectional analysis of 1674 participants from four centers of the Hispanic Community Health Study/Study of Latinos (HCHS/SOL), aged 18 to 74 years old at recruitment. RESULTS Amplicon sequencing of 16S rRNA gene V4 and fungal ITS1 fragments from self-collected stool samples indicate that the host microbiome is determined by sociodemographic and migration-related variables. Those who relocate from Latin America to the USA at an early age have reductions in Prevotella to Bacteroides ratios that persist across the life course. Shannon index of alpha diversity in fungi and bacteria is low in those who relocate to the USA in early life. In contrast, those who relocate to the USA during adulthood, over 45 years old, have high bacterial and fungal diversity and high Prevotella to Bacteroides ratios, compared to USA-born and childhood arrivals. Low bacterial diversity is associated in turn with obesity. Contrasting with prior studies, our study of the Latino population shows increasing Prevotella to Bacteroides ratio with greater obesity. Taxa within Acidaminococcus, Megasphaera, Ruminococcaceae, Coriobacteriaceae, Clostridiales, Christensenellaceae, YS2 (Cyanobacteria), and Victivallaceae are significantly associated with both obesity and earlier exposure to the USA, while Oscillospira and Anaerotruncus show paradoxical associations with both obesity and late-life introduction to the USA. CONCLUSIONS Our analysis of the gut microbiome of Latinos demonstrates unique features that might be responsible for health disparities affecting Hispanics living in the USA.
Collapse
Affiliation(s)
- Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Zheng Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Mykhaylo Usyk
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY USA
| | - Daniela Sotres-Alvarez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC USA
| | - Martha L. Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago College of Medicine, Chicago, IL USA
| | | | - Gregory A. Talavera
- Division of Health Promotion and Behavioral Science, San Diego State University, San Diego, CA USA
| | - Marc D. Gellman
- Department of Psychology, University of Miami, Miami, FL USA
| | - Bharat Thyagarajan
- Division of Molecular Pathology and Genomics, University of Minnesota, Minneapolis, MN USA
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Yoshiki Vázquez-Baeza
- Jacobs School of Engineering, University of California, San Diego, La Jolla, CA USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA USA
| | - Daniel McDonald
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
| | | | - Michael C. Wu
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Kari E. North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC USA
| | - Justin Shaffer
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
| | | | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Carmen R. Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA USA
| | - Robert D. Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY USA
| |
Collapse
|
58
|
Li Y, Alaimo CP, Kim M, Kado NY, Peppers J, Xue J, Wan C, Green PG, Zhang R, Jenkins BM, Vogel CFA, Wuertz S, Young TM, Kleeman MJ. Composition and Toxicity of Biogas Produced from Different Feedstocks in California. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:11569-11579. [PMID: 31479247 PMCID: PMC7608650 DOI: 10.1021/acs.est.9b03003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Biogas is a renewable energy source composed of methane, carbon dioxide, and other trace compounds produced from anaerobic digestion of organic matter. A variety of feedstocks can be combined with different digestion techniques that each yields biogas with different trace compositions. California is expanding biogas production systems to help meet greenhouse gas reduction goals. Here, we report the composition of six California biogas streams from three different feedstocks (dairy manure, food waste, and municipal solid waste). The chemical and biological composition of raw biogas is reported, and the toxicity of combusted biogas is tested under fresh and photochemically aged conditions. Results show that municipal waste biogas contained elevated levels of chemicals associated with volatile chemical products such as aromatic hydrocarbons, siloxanes, and certain halogenated hydrocarbons. Food waste biogas contained elevated levels of sulfur-containing compounds including hydrogen sulfide, mercaptans, and sulfur dioxide. Biogas produced from dairy manure generally had lower concentrations of trace chemicals, but the combustion products had slightly higher toxicity response compared to the other feedstocks. Atmospheric aging performed in a photochemical smog chamber did not strongly change the toxicity (oxidative capacity or mutagenicity) of biogas combustion exhaust.
Collapse
Affiliation(s)
- Yin Li
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Christopher P. Alaimo
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Minji Kim
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Norman Y. Kado
- Department of Environmental Toxicology and Center for Health and the Environment, University of California – Davis, Davis, California 95616, United States
| | - Joshua Peppers
- Department of Biological and Agricultural Engineering, University of California – Davis, Davis, California 95616, United States
| | - Jian Xue
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Chao Wan
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Peter G. Green
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Ruihong Zhang
- Department of Biological and Agricultural Engineering, University of California – Davis, Davis, California 95616, United States
| | - Bryan M. Jenkins
- Department of Biological and Agricultural Engineering, University of California – Davis, Davis, California 95616, United States
| | - Christoph F. A. Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California – Davis, Davis, California 95616, United States
| | - Stefan Wuertz
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technical University, Singapore 637551
| | - Thomas M. Young
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| | - Michael J. Kleeman
- Department of Civil and Environmental Engineering, University of California – Davis, Davis, California 95616, United States
| |
Collapse
|
59
|
Adebowale TO, Yao K, Oso AO. Major cereal carbohydrates in relation to intestinal health of monogastric animals: A review. ACTA ACUST UNITED AC 2019; 5:331-339. [PMID: 31890909 PMCID: PMC6920401 DOI: 10.1016/j.aninu.2019.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/09/2019] [Accepted: 09/05/2019] [Indexed: 01/10/2023]
Abstract
Type, quality, and origin of cereals in diets of poultry and pigs could influence gut microbes and affect their diversity and function, thereby impacting the intestinal function of the monogastric animal. In this review, we focus on the major carbohydrates in cereals that interact directly with gut microbes and lead to the production of key metabolites such as short-chain fatty acids (SCFA), and discuss how cereal fiber impact intestinal health of poultry and pigs. An overview of how the cereals and cereals-derived carbohydrates such as beta-glucans, resistant starch, cellulose, and arabinoxylans could promote intestinal health and reduce the use of in-feed antibiotics in animal production are presented. The metabolic pathway utilized by microbes and the mechanism of action underlying the produced SCFA on intestinal health of monogastric animals is also discussed.
Collapse
Affiliation(s)
- Tolulope O. Adebowale
- University of the Chinese Academy of Sciences, Beijing, 10008, China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China
- Corresponding authors.
| | - Kang Yao
- University of the Chinese Academy of Sciences, Beijing, 10008, China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, 410125, China
- Corresponding authors.
| | - Abimbola O. Oso
- Department of Animal Nutrition, Federal University of Agriculture, Abeokuta, PMB 2240, Nigeria
| |
Collapse
|
60
|
Xiao S, Zhang Z, Chen M, Zou J, Jiang S, Qian D, Duan J. Xiexin Tang ameliorates dyslipidemia in high-fat diet-induced obese rats via elevating gut microbiota-derived short chain fatty acids production and adjusting energy metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:112032. [PMID: 31220598 DOI: 10.1016/j.jep.2019.112032] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/16/2019] [Accepted: 06/16/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional herbal medicine has been taken as a new and effective approach to treat many chronic diseases. Xiexin Tang (XXT), a compound recipe composed of Dahuang (Rheum palmatum L.), Huangqin (Scutellaria baicalensis Georgi) and Huanglian (Coptis chinensis Franch.), has been reported to have hypoglycemic and hypolipidemic effects, but its mechanism remains unclear. Our previous study found that Xiexin Tang markedly ameliorated the composition of the gut microbiota, especially for some short chain fatty acids (SCFAs) producing bacteria, and then notably increased SCFAs production. However, the mechanism of XXT on the fermentation of gut bacteria and further improvement of obesity is not yet clear. AIM OF THE STUDY This study aimed to unravel the molecular mechanism of XXT on the amelioration of obesity. MATERIALS AND METHODS Here, high-fat diet-induced obese rat model was established to investigate the intervention efficacy following oral administration of XXT. Additionally, the expressions of key enzymes of gut microbe-derived SCFAs biosynthesis and key targets in the signaling pathway of energy metabolism were investigated by ELISA and qPCR analysis. RESULTS Results showed that XXT could notably correct lipid metabolism disorders, alleviate systematic inflammation, improve insulin sensitivity and reduce fat accumulation. Additionally, XXT could increase gut microbiota-derived SCFAs-producing capacity by enhancing mRNA levels and activities of SCFA-synthetic key enzymes such as acetate kinase (ACK), methylmalonyl-CoA decarboxylase (MMD), butyryl-CoA: acetate CoA transferase (BUT) and butyrate kinase (BUK), which markedly decreased the adenosine triphosphate (ATP) contents, elevated adenosine diphosphate (ADP) and adenosine monophosphate (AMP) levels and further lowered the energy charge (EC) in obese rats via activating peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)/uncoupling protein-2 (UCP-2) signaling pathway. What's more, XXT could notably ameliorate dyslipidemia via increasing the gene expression of 5'-AMP-activated protein kinase (AMPK) and blocking mammalian target of rapamycin (mTOR) signaling pathway. CONCLUSIONS Taken together, our data provided a novel insight into the role of XXT in losing weight from energy metabolism regulation, which unraveled the molecular mechanism of XXT on the alleviation of dyslipidemia and fat heterotopic accumulation. The study provided useful information for XXT in clinical application to treat obesity.
Collapse
Affiliation(s)
- Suwei Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Zhimiao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Mengjun Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Junfeng Zou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Dawei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| |
Collapse
|
61
|
Chai LJ, Lu ZM, Zhang XJ, Ma J, Xu PX, Qian W, Xiao C, Wang ST, Shen CH, Shi JS, Zheng-Hong X. Zooming in on Butyrate-Producing Clostridial Consortia in the Fermented Grains of Baijiu via Gene Sequence-Guided Microbial Isolation. Front Microbiol 2019; 10:1397. [PMID: 31316481 PMCID: PMC6611424 DOI: 10.3389/fmicb.2019.01397] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/04/2019] [Indexed: 11/13/2022] Open
Abstract
Butyrate, one of the key aroma compounds in Luzhou-flavor baijiu, is synthesized through two alternative pathways: butyrate kinase (buk) and butyryl-CoA: acetate CoA-transferase (but). A lack of knowledge of butyrate-producing microorganisms hinders our ability to understand the flavor formation mechanism of baijiu. Here, temporal dynamics of microbial metabolic profiling in fermented grains (FG) was explored via PICRUSt based on 16S rRNA gene sequences. We found Bacilli and Bacteroidia were the major potential butyrate producers in buk pathway at the beginning of fermentation, while later Clostridia dominated the two pathways. Clone library analysis also revealed that Clostridia (~73% OTUs) was predominant in buk pathway throughout fermentation, followed by Bacilli and Bacteroidia, and but pathway was merely possessed by Clostridia. Afterward, Clostridia-specific 16S rRNA gene sequencing demonstrated Clostridium might be the major butyrate-producing genus in two pathways, which was subsequently evaluated using culture approach. Seventeen Clostridium species were isolated from FG based on 16S rRNA gene sequence-guided medium prediction method. Profiles of short-chain fatty acids and but and buk genes in these species demonstrated phylogenetic and functional diversities of butyrate-producing Clostridium in FG. These findings add to illustrate the diversity of potential butyrate producers during brewing and provide a workflow for targeting functional microbes in complex microbial community.
Collapse
Affiliation(s)
- Li-Juan Chai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, Jiangnan University, Wuxi, China
| | - Zhen-Ming Lu
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, Jiangnan University, Wuxi, China.,School of Pharmaceutical Science, Jiangnan University, Wuxi, China
| | - Xiao-Juan Zhang
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center of Solid-State Brewing, Luzhou, China
| | - Jian Ma
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Peng-Xiang Xu
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Wei Qian
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Chen Xiao
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Song-Tao Wang
- National Engineering Research Center of Solid-State Brewing, Luzhou, China
| | - Cai-Hong Shen
- National Engineering Research Center of Solid-State Brewing, Luzhou, China
| | - Jin-Song Shi
- School of Pharmaceutical Science, Jiangnan University, Wuxi, China
| | - Xu Zheng-Hong
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.,National Engineering Research Center of Solid-State Brewing, Luzhou, China
| |
Collapse
|
62
|
Villanueva-Millán MJ, Pérez-Matute P, Recio-Fernández E, Lezana Rosales JM, Oteo JA. Characterization of gut microbiota composition in HIV-infected patients with metabolic syndrome. J Physiol Biochem 2019; 75:299-309. [PMID: 30924020 DOI: 10.1007/s13105-019-00673-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/06/2019] [Indexed: 12/26/2022]
Abstract
The presence of metabolic syndrome (MS) per se or its separated components in HIV-infected patients contributes to an accelerated aging and increased cardiovascular risk. Gut microbiota (GM) dysbiosis has been linked with chronic inflammation associated with MS in a general non-infected population. However, no studies concerning GM have been performed in HIV-infected patients with MS. The aim of this study was to analyze bacterial translocation, inflammation, and GM composition in HIV-infected patients with and without MS. A total of 51 HIV-infected patients were recruited and classified according to the presence of MS (40 patients without MS and 11 with MS). Markers of bacterial translocation, inflammation, and cardiovascular risk were measured and GM was analyzed using 16S rRNA gene deep sequencing. No differences were observed among both HIV-infected groups in the bacterial translocation markers LBP and sCD14. A tendency to increase the inflammatory markers IL-6 (p = 0.069) and MCP-1 (p = 0.067) was observed in those patients suffering from MS. An increase in the cardiovascular risk markers PAI-1 (p = 0.007) and triglycerides/HDL cholesterol ratio (p < 0.0001) was also found in the MS group. No significant changes were observed at phylum level although a decrease in the abundance of seven genera and seven bacterial species, including some anti-inflammatory bacteria, was observed in HIV-infected patients with MS. To summarize, the presence of MS was not accompanied by major changes in GM, although the reduction observed in some anti-inflammatory bacteria may be clinically useful to develop strategies to minimize inflammation and its future deleterious consequences in these HIV-infected patients.
Collapse
Affiliation(s)
- María Jesús Villanueva-Millán
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain
| | - Patricia Pérez-Matute
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain.
| | - Emma Recio-Fernández
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain
| | - José-Miguel Lezana Rosales
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain
| | - José-Antonio Oteo
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain.,Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, La Rioja, Spain
| |
Collapse
|
63
|
Profiling the Clostridia with butyrate-producing potential in the mud of Chinese liquor fermentation cellar. Int J Food Microbiol 2019; 297:41-50. [PMID: 30878841 DOI: 10.1016/j.ijfoodmicro.2019.02.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 01/03/2023]
Abstract
Butyrate and its derivates pertain to the key aroma contributors of strong-flavour baijiu, a kind of Chinese liquors, that is produced from grains by solid-state multispecies anaerobic fermentation in a mud cellar. Microbes inhabiting in the fermentation pit mud largely determines baijiu's flavour and quality. In order to shed light on the microbial functional groups driving butyrate production in pit mud, clone library analysis was firstly performed and the results demonstrated that Clostridia (relative abundance: 50%) and Bacilli (37%) were major groups possessing butyrate kinase (buk) pathway and Clostridia (98%) dominated butyryl-CoA:acetate CoA-transferase (but) pathway. According to Clostridial specific-16S rRNA gene sequencing analysis, we found the resilience character of Clostridial community in pit mud. Amongst Clostridial groups, 32.0% of the sequences were grouped into Clostridiales incertae sedis, followed by Heliobacteriaceae (18.3%) and Clostridiaceae 1 (8.4%). Moreover, Hydrogenispora, Sedimentibacter and Clostridium were the top three abundant genera. Relative abundance of Hydrogenispora was higher in the late days of fermentation, while Sedimentibacter exhibited higher proportion in the early days. Different from the previous studies using universal bacterial primer sets, Hydrogenispora was first reported as one dominant genus in pit mud. As for the reported potential butyrate producer Clostridium, nineteen species were obtained and ten of them were first isolated from the pit mud. Amongst them, buk was identified in eleven species by PCR analysis, while but was identified in the other seven, indicating the species-specific butyrate synthesis pathways of Clostridium. This study provides a perspective on targeting and isolating specific functional microbes in baijiu microbiota with the gene sequence-based medium prediction method.
Collapse
|
64
|
Grate JW, Liu B, Kelly RT, Anheier NC, Schmidt TM. Microfluidic Sensors with Impregnated Fluorophores for Simultaneous Imaging of Spatial Structure and Chemical Oxygen Gradients. ACS Sens 2019; 4:317-325. [PMID: 30609370 DOI: 10.1021/acssensors.8b00924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Interior surfaces of polystyrene microfluidic structures were impregnated with the oxygen sensing dye Pt(II) tetra(pentafluorophenyl)porphyrin (PtTFPP) using a solvent-induced fluorophore impregnation (SIFI) method. Using this technique, microfluidic oxygen sensors are obtained that enable simultaneous imaging of both chemical oxygen gradients and the physical structure of the microfluidic interior. A gentle method of fluorophore impregnation using acetonitrile solutions of PtTFPP at 50 °C was developed leading to a 10-μm-deep region containing fluorophore. This region is localized at the surface to sense oxygen in the interior fluid during use. Regions of the device that do not contact the interior fluid pathways lack fluorophores and are dark in fluorescent imaging. The technique was demonstrated on straight microchannel and pore network devices, the latter having pillars of 300 μm diameter spaced center to center at 340 μm providing pore throats of 40 μm. Sensing within channels or pores and imaging across the pore network devices were performed using a Lambert LIFA-P frequency domain fluorescence lifetime imaging system on a Leica microscope platform. Calibrations of different devices prepared by the SIFI method were indistinguishable. Gradient imaging showed fluorescent regions corresponding to the fluid pore network, dark pillars, and fluorescent lifetime varying across the gradient, thus providing both physical and chemical imaging. More generally, the SIFI technique can impregnate the interior surfaces of other polystyrene containers, such as cuvettes or cell and tissue culture containers, to enable sensing of interior conditions.
Collapse
Affiliation(s)
- Jay W. Grate
- Pacific Northwest
National Laboratory, P.O. Box 999, Richland, Washington 99352, United States
| | - Bingwen Liu
- Pacific Northwest
National Laboratory, P.O. Box 999, Richland, Washington 99352, United States
| | - Ryan T. Kelly
- Pacific Northwest
National Laboratory, P.O. Box 999, Richland, Washington 99352, United States
| | - Norman C. Anheier
- Pacific Northwest
National Laboratory, P.O. Box 999, Richland, Washington 99352, United States
| | - Thomas M. Schmidt
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
65
|
Fontana A, Kougias PG, Treu L, Kovalovszki A, Valle G, Cappa F, Morelli L, Angelidaki I, Campanaro S. Microbial activity response to hydrogen injection in thermophilic anaerobic digesters revealed by genome-centric metatranscriptomics. MICROBIOME 2018; 6:194. [PMID: 30368244 PMCID: PMC6204281 DOI: 10.1186/s40168-018-0583-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/18/2018] [Indexed: 05/10/2023]
Abstract
BACKGROUND The expansion of renewable energy produced by windmills and photovoltaic panels has generated a considerable electricity surplus, which can be utilized in water electrolysis systems for hydrogen production. The resulting hydrogen can then be funneled to anaerobic digesters for biogas upgrading (biomethanation) purposes (power-to-methane) or to produce high value-added compounds such as short-chain fatty acids (power-to-chemicals). Genome-centric metagenomics and metatranscriptomic analyses were performed to better understand the metabolic dynamics associated with H2 injection in two different configurations of anaerobic digesters treating acidic wastes, specifically cheese manufacturing byproducts. These approaches revealed the key-genes involved in methanation and carbon fixation pathways at species level. RESULTS The biogas upgrading process in the single-stage configuration increased the CH4 content by 7%. The dominant methanogenic species responsible for the upregulation of the hydrogenotrophic pathway in this reactor was Methanothermobacter wolfeii UC0008. In the two-stage configuration, H2 injection induced an upregulation of CO2 fixation pathways producing short-chain fatty acids, mainly acetate and butyrate. In this configuration, the abundant species Anaerobaculum hydrogeniformans UC0046 and Defluviitoga tunisiensis UC0050 primarily upregulated genes related to electron transport chains, suggesting putative syntrophisms with hydrogen scavenger microbes. Interestingly, Tepidanaerobacter acetatoxydans UC0018 did not act as an acetate-oxidizer in either reactor configurations, and instead regulated pathways involved in acetate production and uptake. A putative syntrophic association between Coprothermobacter proteolyticus UC0011 and M. wolfeii UC0008 was proposed in the two-stage reactor. In order to support the transcriptomic findings regarding the hydrogen utilization routes, an advanced bioconversion model was adapted for the simulation of the single- and two-stage reactor setups. CONCLUSIONS This is the first study investigating biogas reactor metatranscriptome dynamics following hydrogen injection for biomethanation and carbon fixation to short-chain fatty acids purposes. The same microbes showed different patterns of metabolic regulation in the two reactor configurations. It was observed an effect of the specialized acidogenic reactor on the overall microbial consortium composition and activity in the two-stage digester. There were also suggested the main species responsible for methanation, short-chain fatty acids production, and electron transport chain mechanisms, in both reactor configurations.
Collapse
Affiliation(s)
- Alessandra Fontana
- Department for Sustainable Food Process, DiSTAS, Catholic University of the Sacred Heart, 29122, Piacenza, Italy
- Department of Environmental Engineering, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Panagiotis G Kougias
- Department of Environmental Engineering, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Laura Treu
- Department of Environmental Engineering, Technical University of Denmark, 2800, Kongens Lyngby, Denmark.
| | - Adam Kovalovszki
- Department of Environmental Engineering, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Giorgio Valle
- Department of Biology, University of Padua, 35131, Padua, Italy
| | - Fabrizio Cappa
- Department for Sustainable Food Process, DiSTAS, Catholic University of the Sacred Heart, 29122, Piacenza, Italy
| | - Lorenzo Morelli
- Department for Sustainable Food Process, DiSTAS, Catholic University of the Sacred Heart, 29122, Piacenza, Italy
| | - Irini Angelidaki
- Department of Environmental Engineering, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | | |
Collapse
|
66
|
Small Intestine Microbiota Regulate Host Digestive and Absorptive Adaptive Responses to Dietary Lipids. Cell Host Microbe 2018; 23:458-469.e5. [PMID: 29649441 DOI: 10.1016/j.chom.2018.03.011] [Citation(s) in RCA: 374] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 11/15/2017] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
The gut microbiota play important roles in lipid metabolism and absorption. However, the contribution of the small bowel microbiota of mammals to these diet-microbe interactions remains unclear. We determine that germ-free (GF) mice are resistant to diet-induced obesity and malabsorb fat with specifically impaired lipid digestion and absorption within the small intestine. Small bowel microbes are essential for host adaptation to dietary lipid changes by regulating gut epithelial processes involved in their digestion and absorption. In addition, GF mice conventionalized with high-fat diet-induced jejunal microbiota exhibit increased lipid absorption even when fed a low-fat diet. Conditioned media from specific bacterial strains directly upregulate lipid absorption genes in murine proximal small intestinal epithelial organoids. These findings indicate that proximal gut microbiota play key roles in host adaptability to dietary lipid variations through mechanisms involving both the digestive and absorptive phases and that these functions may contribute to conditions of over- and undernutrition.
Collapse
|
67
|
Passmore IJ, Letertre MPM, Preston MD, Bianconi I, Harrison MA, Nasher F, Kaur H, Hong HA, Baines SD, Cutting SM, Swann JR, Wren BW, Dawson LF. Para-cresol production by Clostridium difficile affects microbial diversity and membrane integrity of Gram-negative bacteria. PLoS Pathog 2018; 14:e1007191. [PMID: 30208103 PMCID: PMC6135563 DOI: 10.1371/journal.ppat.1007191] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/29/2018] [Indexed: 11/18/2022] Open
Abstract
Clostridium difficile is a Gram-positive spore-forming anaerobe and a major cause of antibiotic-associated diarrhoea. Disruption of the commensal microbiota, such as through treatment with broad-spectrum antibiotics, is a critical precursor for colonisation by C. difficile and subsequent disease. Furthermore, failure of the gut microbiota to recover colonisation resistance can result in recurrence of infection. An unusual characteristic of C. difficile among gut bacteria is its ability to produce the bacteriostatic compound para-cresol (p-cresol) through fermentation of tyrosine. Here, we demonstrate that the ability of C. difficile to produce p-cresol in vitro provides a competitive advantage over gut bacteria including Escherichia coli, Klebsiella oxytoca and Bacteroides thetaiotaomicron. Metabolic profiling of competitive co-cultures revealed that acetate, alanine, butyrate, isobutyrate, p-cresol and p-hydroxyphenylacetate were the main metabolites responsible for differentiating the parent strain C. difficile (630Δerm) from a defined mutant deficient in p-cresol production. Moreover, we show that the p-cresol mutant displays a fitness defect in a mouse relapse model of C. difficile infection (CDI). Analysis of the microbiome from this mouse model of CDI demonstrates that colonisation by the p-cresol mutant results in a distinctly altered intestinal microbiota, and metabolic profile, with a greater representation of Gammaproteobacteria, including the Pseudomonales and Enterobacteriales. We demonstrate that Gammaproteobacteria are susceptible to exogenous p-cresol in vitro and that there is a clear divide between bacterial Phyla and their susceptibility to p-cresol. In general, Gram-negative species were relatively sensitive to p-cresol, whereas Gram-positive species were more tolerant. This study demonstrates that production of p-cresol by C. difficile has an effect on the viability of intestinal bacteria as well as the major metabolites produced in vitro. These observations are upheld in a mouse model of CDI, in which p-cresol production affects the biodiversity of gut microbiota and faecal metabolite profiles, suggesting that p-cresol production contributes to C. difficile survival and pathogenesis.
Collapse
Affiliation(s)
- Ian J. Passmore
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Mark D. Preston
- Bioinformatics and Next Generation sequencing core facility, National Institute for Biological Standards and Control South Mimms, Potters Bar, United Kingdom
| | - Irene Bianconi
- Department of Biomedical Sciences, Royal Holloway University of London, Egham, United Kingdom
| | - Mark A. Harrison
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fauzy Nasher
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Harparkash Kaur
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Huynh A. Hong
- Department of Biomedical Sciences, Royal Holloway University of London, Egham, United Kingdom
| | - Simon D. Baines
- Department of Biological and Environmental Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Simon M. Cutting
- Department of Biomedical Sciences, Royal Holloway University of London, Egham, United Kingdom
| | - Jonathan R. Swann
- Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Brendan W. Wren
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Lisa F. Dawson
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
68
|
Nguyen TTT, Fujimura Y, Mimura I, Fujii Y, Nguyen NL, Arakawa K, Morita H. Cultivable butyrate-producing bacteria of elderly Japanese diagnosed with Alzheimer’s disease. J Microbiol 2018; 56:760-771. [DOI: 10.1007/s12275-018-8297-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/04/2018] [Accepted: 07/04/2018] [Indexed: 12/16/2022]
|
69
|
Metatranscriptome analysis of the microbial fermentation of dietary milk proteins in the murine gut. PLoS One 2018; 13:e0194066. [PMID: 29664912 PMCID: PMC5903625 DOI: 10.1371/journal.pone.0194066] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 02/23/2018] [Indexed: 01/22/2023] Open
Abstract
Undigestible food ingredients are converted by the microbiota into a large range of metabolites, predominated by short chain fatty acids (SCFA). These microbial metabolites are subsequently available for absorption by the host mucosa and can serve as an energy source. Amino acids fermentation by the microbiota expands the spectrum of fermentation end-products beyond acetate, propionate and butyrate, to include in particular branched-SCFA. Here the long-term effects of high protein-diets on microbial community composition and functionality in mice were analyzed. Determinations of the microbiota composition using phylogenetic microarray (MITChip) technology were complemented with metatranscriptome and SCFA analyses to obtain insight in in situ expression of protein fermentation pathways and the phylogenetic groups involved. High protein diets led to increased luminal concentrations of branched-SCFA, in accordance with protein fermentation in the gut. Bacteria dominantly participating in protein catabolism belonged to the Lachnospiraceae, Erysipelotrichaceae and Clostridiaceae families in both normal- and high- protein diet regimes. This study identifies the microbial groups involved in protein catabolism in the intestine and underpins the value of in situ metatranscriptome analyses as an approach to decipher locally active metabolic networks and pathways as a function of the dietary regime, as well as the phylogeny of the microorganisms executing them.
Collapse
|
70
|
Ong HS, Yim HCH. Microbial Factors in Inflammatory Diseases and Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1024:153-174. [PMID: 28921469 DOI: 10.1007/978-981-10-5987-2_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The intestinal microbes form a symbiotic relationship with their human host to harvest energy for themselves and their host and to shape the immune system of their host. However, alteration of this relationship, which is named as a dysbiosis, has been associated with the development of different inflammatory diseases and cancers. It is found that metabolites, cellular components, and virulence factors derived from the gut microbiota interact with the host locally or systemically to modulate the dysbiosis and the development of these diseases. In this book chapter, we discuss the role of these microbial factors in regulating the host signaling pathways, the composition and load of the gut microbiota, the co-metabolism of the host and the microbiota, the host immune system, and physiology. In particular, we highlight how each microbial factor can contribute in the manifestation of many diseases such as cancers, Inflammatory Bowel Diseases, obesity, type-2 diabetes, non-alcoholic fatty liver diseases, nonalcoholic steatohepatitis, and cardiovascular diseases.
Collapse
Affiliation(s)
- Hong Sheng Ong
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Acton, Canberra, 2601, ACT, Australia
| | - Howard Chi Ho Yim
- Department of Medicine, St George & Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Australia, Level 2 Clinical Sciences (WR Pitney) Building, St George Hospital, Short St, Kogarah, NSW, 2217, Australia.
| |
Collapse
|
71
|
Dietary butyrate glycerides modulate intestinal microbiota composition and serum metabolites in broilers. Sci Rep 2018; 8:4940. [PMID: 29563518 PMCID: PMC5862971 DOI: 10.1038/s41598-018-22565-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/13/2018] [Indexed: 12/21/2022] Open
Abstract
Butyrate can modulate the immune response and energy expenditure of animals and enhance intestinal health. The present study investigated changes in the intestinal microbiota composition and serum metabolites of young broilers in response to 3,000 ppm butyrate in the form of butyrate glycerides (BG) via pyrosequencing of bacterial 16S rRNA genes and nuclear magnetic resonance (NMR). The dietary treatment did not affect the alpha diversity of intestinal microbiota, but altered its composition. Thirty-nine key operational taxonomic units (OTUs) in differentiating cecal microbiota community structures between BG treated and untreated chickens were also identified. Bifidobacterium was, in particular, affected by the dietary treatment significantly, showing an increase in not only the abundance (approximately 3 fold, P ≤ 0.05) but also the species diversity. The (NMR)-based analysis revealed an increase in serum concentrations of alanine, low-density and very low-density lipoproteins, and lipids (P ≤ 0.05) by BG. More interestingly, the dietary treatment also boosted (P ≤ 0.05) serum concentrations of bacterial metabolites, including choline, glycerophosphorylcholine, dimethylamine, trimethylamine, trimethylamine-N-oxide, lactate, and succinate. In conclusion, the data suggest the modulation of intestinal microbiota and serum metabolites by BG dietary treatment and potential contribution of intestinal bacteria to lipid metabolism/energy homeostasis in broilers.
Collapse
|
72
|
Šket R, Debevec T, Kublik S, Schloter M, Schoeller A, Murovec B, Vogel Mikuš K, Makuc D, Pečnik K, Plavec J, Mekjavić IB, Eiken O, Prevoršek Z, Stres B. Intestinal Metagenomes and Metabolomes in Healthy Young Males: Inactivity and Hypoxia Generated Negative Physiological Symptoms Precede Microbial Dysbiosis. Front Physiol 2018; 9:198. [PMID: 29593560 PMCID: PMC5859311 DOI: 10.3389/fphys.2018.00198] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/23/2018] [Indexed: 12/26/2022] Open
Abstract
We explored the metagenomic, metabolomic and trace metal makeup of intestinal microbiota and environment in healthy male participants during the run-in (5 day) and the following three 21-day interventions: normoxic bedrest (NBR), hypoxic bedrest (HBR) and hypoxic ambulation (HAmb) which were carried out within a controlled laboratory environment (circadian rhythm, fluid and dietary intakes, microbial bioburden, oxygen level, exercise). The fraction of inspired O2 (FiO2) and partial pressure of inspired O2 (PiO2) were 0.209 and 133.1 ± 0.3 mmHg for the NBR and 0.141 ± 0.004 and 90.0 ± 0.4 mmHg (~4,000 m simulated altitude) for HBR and HAmb interventions, respectively. Shotgun metagenomes were analyzed at various taxonomic and functional levels, 1H- and 13C -metabolomes were processed using standard quantitative and human expert approaches, whereas metals were assessed using X-ray fluorescence spectrometry. Inactivity and hypoxia resulted in a significant increase in the genus Bacteroides in HBR, in genes coding for proteins involved in iron acquisition and metabolism, cell wall, capsule, virulence, defense and mucin degradation, such as beta-galactosidase (EC3.2.1.23), α-L-fucosidase (EC3.2.1.51), Sialidase (EC3.2.1.18), and α-N-acetylglucosaminidase (EC3.2.1.50). In contrast, the microbial metabolomes, intestinal element and metal profiles, the diversity of bacterial, archaeal and fungal microbial communities were not significantly affected. The observed progressive decrease in defecation frequency and concomitant increase in the electrical conductivity (EC) preceded or took place in absence of significant changes at the taxonomic, functional gene, metabolome and intestinal metal profile levels. The fact that the genus Bacteroides and proteins involved in iron acquisition and metabolism, cell wall, capsule, virulence and mucin degradation were enriched at the end of HBR suggest that both constipation and EC decreased intestinal metal availability leading to modified expression of co-regulated genes in Bacteroides genomes. Bayesian network analysis was used to derive the first hierarchical model of initial inactivity mediated deconditioning steps over time. The PlanHab wash-out period corresponded to a profound life-style change (i.e., reintroduction of exercise) that resulted in stepwise amelioration of the negative physiological symptoms, indicating that exercise apparently prevented the crosstalk between the microbial physiology, mucin degradation and proinflammatory immune activities in the host.
Collapse
Affiliation(s)
- Robert Šket
- Group for Microbiology and Microbial Biotechnology, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Debevec
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute, Ljubljana, Slovenia.,Faculty of Sport, University of Ljubljana, Ljubljana, Slovenia
| | - Susanne Kublik
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Schloter
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Anne Schoeller
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Boštjan Murovec
- Machine Vision Laboratory, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Vogel Mikuš
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Damjan Makuc
- Slovenian NMR Centre, National Institute of Chemistry, Ljubljana, Slovenia
| | - Klemen Pečnik
- Slovenian NMR Centre, National Institute of Chemistry, Ljubljana, Slovenia
| | - Janez Plavec
- Slovenian NMR Centre, National Institute of Chemistry, Ljubljana, Slovenia
| | - Igor B Mekjavić
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Ola Eiken
- Department of Environmental Physiology, Swedish Aerospace Physiology Centre, Royal Institute of Technology, Stockholm, Sweden
| | - Zala Prevoršek
- Group for Genetics, Animal Biotechnology and Immunology, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Blaž Stres
- Group for Microbiology and Microbial Biotechnology, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia.,Center for Clinical Neurophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
73
|
Miyoshi J, Qiao Y, Chang EB. The role of the intestinal microbiota in the pathogenesis and treatment of inflammatory bowel diseases. SEMINARS IN COLON AND RECTAL SURGERY 2018. [DOI: 10.1053/j.scrs.2017.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
74
|
Fischer AJ, Song Y, He Z, Haines DM, Guan LL, Steele MA. Effect of delaying colostrum feeding on passive transfer and intestinal bacterial colonization in neonatal male Holstein calves. J Dairy Sci 2018; 101:3099-3109. [PMID: 29397179 DOI: 10.3168/jds.2017-13397] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
Abstract
The objective of this study was to investigate the effect of time of first colostrum feeding on the passive transfer of IgG and on bacterial colonization in the intestine of neonatal dairy calves. Twenty-seven male Holstein calves were randomly assigned to 1 of 3 treatments at birth: calves were fed colostrum at 45 min (0 h, n = 9), 6 h (n = 9), or 12 h after birth (n = 9). Calves were fed pooled, heat-treated colostrum (62 g of IgG/L) at their respective feeding times at 7.5% of birth body weight and fed milk replacer at 2.5% of birth body weight per meal every 6 h thereafter. Blood samples were taken every 3 h using a jugular catheter and were analyzed for determination of serum IgG by radial immunodiffusion. At 51 h after birth, calves were euthanized for collection of tissue and digesta of the distal jejunum, ileum, and colon. Quantitative real-time PCR was used to estimate the prevalence of Bifidobacterium spp., Lactobacillus spp., Fecalibacterium prausnitzii, Clostridium cluster XIVa, and total Escherichia coli. Delaying colostrum feeding by 6 h (35.6 ± 1.88%) and 12 h (35.1 ± 3.15%) decreased the maximum apparent efficiency of absorption of IgG compared with feeding colostrum immediately after birth (51.8 ± 4.18%) and delayed the time to maximum serum IgG concentration (24 h vs. 15 h, respectively). Moreover, 12-h calves tended to have a lower prevalence of Bifidobacterium spp. (0.12 ± 0.017%) and Lactobacillus spp. (0.07 ± 0.019%) associated with the colon mucosa compared with 0-h calves (1.24 ± 0.648% and 0.26 ± 0.075%, respectively). In addition, 6-h (0.26 ± 0.124%) and 12-h (0.49 ± 0.233%) calves had a lower prevalence of total E. coli associated with ileum mucosa compared with 0-h calves (1.20 ± 0.458%). These findings suggest that delaying colostrum feeding within 12 h of life decreases the passive transfer of IgG and may delay the colonization of bacteria in the intestine, possibly leaving the calf vulnerable to infections during the preweaning period.
Collapse
Affiliation(s)
- A J Fischer
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5
| | - Y Song
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5
| | - Z He
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5
| | - D M Haines
- Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 5B4; The Saskatoon Colostrum Co. Ltd., Saskatoon, Saskatchewan, Canada S7K 6A2
| | - L L Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5
| | - M A Steele
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5.
| |
Collapse
|
75
|
Mushroom polysaccharides from Ganoderma lucidum and Poria cocos reveal prebiotic functions. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.046] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
76
|
Bircher L, Schwab C, Geirnaert A, Lacroix C. Cryopreservation of artificial gut microbiota produced with in vitro fermentation technology. Microb Biotechnol 2018; 11:163-175. [PMID: 28980453 PMCID: PMC5743790 DOI: 10.1111/1751-7915.12844] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/29/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Interest in faecal microbiota transplantation (FMT) has increased as therapy for intestinal diseases, but safety issues limit its widespread use. Intestinal fermentation technology (IFT) can produce controlled, diverse and metabolically active 'artificial' colonic microbiota as potential alternative to common FMT. However, suitable processing technology to store this artificial microbiota is lacking. In this study, we evaluated the impact of the two cryoprotectives, glycerol (15% v/v) and inulin (5% w/v) alone and in combination, in preserving short-chain fatty acid formation and recovery of major butyrate-producing bacteria in three artificial microbiota during cryopreservation for 3 months at -80°C. After 24 h anaerobic fermentation of the preserved microbiota, butyrate and propionate production were maintained when glycerol was used as cryoprotectant, while acetate and butyrate were formed more rapidly with glycerol in combination with inulin. Glycerol supported cryopreservation of the Roseburia spp./Eubacterium rectale group, while inulin improved the recovery of Faecalibacterium prausnitzii. Eubacterium hallii growth was affected minimally by cryopreservation. Our data indicate that butyrate producers, which are key organisms for gut health, can be well preserved with glycerol and inulin during frozen storage. This is of high importance if artificially produced colonic microbiota is considered for therapeutic purposes.
Collapse
Affiliation(s)
- Lea Bircher
- Laboratory of Food BiotechnologyInstitute of Food, Nutrition and HealthETH ZürichSchmelzbergstrasse 78092ZürichSwitzerland
| | - Clarissa Schwab
- Laboratory of Food BiotechnologyInstitute of Food, Nutrition and HealthETH ZürichSchmelzbergstrasse 78092ZürichSwitzerland
| | - Annelies Geirnaert
- Laboratory of Food BiotechnologyInstitute of Food, Nutrition and HealthETH ZürichSchmelzbergstrasse 78092ZürichSwitzerland
| | - Christophe Lacroix
- Laboratory of Food BiotechnologyInstitute of Food, Nutrition and HealthETH ZürichSchmelzbergstrasse 78092ZürichSwitzerland
| |
Collapse
|
77
|
Doo EH, Chassard C, Schwab C, Lacroix C. Effect of dietary nucleosides and yeast extracts on composition and metabolic activity of infant gut microbiota in PolyFermS colonic fermentation models. FEMS Microbiol Ecol 2017; 93:3934649. [PMID: 28854667 DOI: 10.1093/femsec/fix088] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Nucleotides (NT) and nucleosides (NS) are added to infant formula to mimic the content of breast milk, but little is known about their impact on infant gut microbiota. In this study, we tested the effect of NS and of yeast extracts (YE) with different NT content using PolyFermS continuous fermentation models mimicking formula-fed, healthy and enteropathogen-contaminated infant gut microbiota. Microbiota composition, short-chain fatty acid (SCFA) formation and gene expression were determined. NS, and to a larger extend YE modulated microbiota composition and increased metabolic activity in both models. Anaerococcus, Peptoniphilus, Fusobacterium, Lactobacillus/Pediococcus/Leuconostoc and Veillonella were enhanced when YE and/or NS were added. The production of SCFA increased with the level of supplied NT equivalents. Addition of NS and YE reduced colonization of Salmonella compared to control periods. Gene expression analysis confirmed taxonomical changes and indicated functional responses to YE. Transcripts related to NT and sulfur metabolism and iron acquisition increased while biosynthesis of co-factors and vitamins decreased after YE addition. Elevated butyrate formation correlated with increased transcripts encoding key enzymes of the two major butyrate synthesis pathways. Our results uncover a strong dose-dependent modulation of NS and YE on infant gut microbiota composition and metabolic activity.
Collapse
Affiliation(s)
- Eun-Hee Doo
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH-Zurich, 8092 Zurich, Switzerland
| | - Christophe Chassard
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH-Zurich, 8092 Zurich, Switzerland.,Université Clermont Auvergne, INRA, UMRF, F-15000 Aurillac, France
| | - Clarissa Schwab
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH-Zurich, 8092 Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH-Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
78
|
The response of canine faecal microbiota to increased dietary protein is influenced by body condition. BMC Vet Res 2017; 13:374. [PMID: 29202841 PMCID: PMC5716228 DOI: 10.1186/s12917-017-1276-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 11/16/2017] [Indexed: 01/06/2023] Open
Abstract
Background High protein diets shift the faecal microbiota into a more unfavourable composition in obese humans. In lean dogs, higher protein consumption is accompanied with increased production of putrefactive fermentation products, whereas obese dogs have a different gut microbiota compared to lean dogs. Still, the impact of high dietary protein on gut microbiota in obese dogs remains unclear. The aim of this study was to investigate faecal microbial changes in lean and obese dogs in response to two different levels of dietary protein. Six healthy lean and six obese Beagles were fed a high protein diet (HP) and a low protein diet (LP) for 28 days each in a crossover design. Denaturing gradient gel electrophoresis and quantitative PCR were performed on faecal samples for microbial profiling. Plasma acylcarnitine and fermentation metabolites were measured. Results Dogs fed HP had higher concentrations of protein fermentation metabolites including faecal ammonia, isovalerate, isobutyrate, phenol, indole, serum indoxyl sulphate and plasma 3-OH isovalerylcarnitine compared to dogs fed LP, whereas no changes in faecal concentrations of acetate and butyrate were observed. The abundances of clostridial clusters IV and XIVa, covering the majority of butyrate-producing bacteria, and of the butyrate kinase gene, one of the terminal genes of the butyrate synthesis pathway were higher in dogs on HP compared to LP. Significant interactions between diet and body condition were found for the abundance of Firmicutes, Lactobacillus and clostridial cluster I. The similarity coefficient of faecal microbiota between the two diets was smaller in obese dogs than in lean dogs. Conclusions High protein diet increased the abundance and activity of butyrate-producing bacteria in Beagles independent of the body condition. In addition, increasing dietary protein content had a greater overall impact on faecal microbiota in obese compared to lean dogs. Electronic supplementary material The online version of this article (10.1186/s12917-017-1276-0) contains supplementary material, which is available to authorized users.
Collapse
|
79
|
Co-supplementation of isomalto-oligosaccharides potentiates metabolic health benefits of polyphenol-rich cranberry extract in high fat diet-fed mice via enhanced gut butyrate production. Eur J Nutr 2017; 57:2897-2911. [PMID: 29127476 DOI: 10.1007/s00394-017-1561-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE Cranberries are a rich source of polyphenolic antioxidants. Purified sugars or artificial sweeteners are being added to cranberry-based food products to mask tartness. Refined sugar and artificial sweeteners intake modulate gut microbiota and result in metabolic complications. We evaluated effects of isomalto-oligosaccharides (IMOs; sweet tasting non-digestible oligosaccharides) with cranberry extract (CRX) on high fat diet (HFD)-induced metabolic alterations in mice. METHODS Male Swiss albino mice were fed normal chow or HFD (58% fat kcal), and were administered either CRX (200 mg/kg) alone or in combination with IMOs (1 g/kg). Cecal short-chain fatty acids, abundances of selected (1) butyrate producing, (2) metabolically beneficial, and (3) selective lipopolysaccharides producing gram negative gut bacteria were studied. Further, gut-related histological, biochemical, genomic changes along with circulating pro-/anti-inflammatory markers and systemic obesity-associated metabolic changes were studied. RESULTS Co-supplementation of CRX and IMOs significantly improved cecal SCFAs, especially butyrate levels, selected butyrate-producing bacteria (clostridial cluster XIVa bacteria) and butyrate kinase expression in HFD-fed mice. The combination also significantly improved gut beneficial bacterial abundance, gut histology and related changes (colon mucin production, gut permeability) as compared to individual agents. It also prevented HFD-induced systemic and tissue inflammation, glucose intolerance and systemic obesity-associated metabolic changes in adipose tissue and liver. The combination of CRX and IMOs appeared more effective in the prevention of HFD-induced gut derangements. CONCLUSION Combination of CRX and IMOs could be advantageous for normalization of metabolic alterations seen in diet-induced obesity via beneficial modulation of gastrointestinal health.
Collapse
|
80
|
Burbach K, Strang EJP, Mosenthin R, Camarinha-Silva A, Seifert J. Porcine intestinal microbiota is shaped by diet composition based on rye or triticale. J Appl Microbiol 2017; 123:1571-1583. [PMID: 28960763 DOI: 10.1111/jam.13595] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/12/2017] [Accepted: 09/17/2017] [Indexed: 01/03/2023]
Abstract
AIMS The present study aimed to compare the microbiota composition from pigs fed different cereal grain types, either rye or triticale, as sole energy source. METHODS AND RESULTS Ileal digesta and faeces were sampled from eight pigs of each experiment. Illumina amplicon sequencing of the 16S rRNA gene was used to analyse the microbiota. Concentrations of short-chain fatty acids and ammonia were determined from faecal samples. The grain type revealed significant alterations in the overall microbiota structure. The rye-based diet was associated with an increased abundance of Lactobacillus in ileal digesta and Streptococcus in faeces and significantly higher concentrations of faecal short-chain fatty acids and ammonia compared to triticale. However, triticale significantly promoted the abundance of Streptococcus in ileal digesta and Clostridium sensu stricto in faeces. CONCLUSIONS Diets based on rye or triticale affect varying intestinal microbiota, both of taxonomical and metabolic structure, with rye indicating an enhanced saccharolytic potential and triticale a more cellulolytic potential. SIGNIFICANCE AND IMPACT OF THE STUDY Nutrient composition of rye and triticale are attractive for porcine nutrition. Both cereal grains show varying stimuli on the microbiota composition and microbial products of the ileum and faeces.
Collapse
Affiliation(s)
- K Burbach
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - E J P Strang
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - R Mosenthin
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - A Camarinha-Silva
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - J Seifert
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
81
|
Tian Y, Nichols RG, Cai J, Patterson AD, Cantorna MT. Vitamin A deficiency in mice alters host and gut microbial metabolism leading to altered energy homeostasis. J Nutr Biochem 2017; 54:28-34. [PMID: 29227833 DOI: 10.1016/j.jnutbio.2017.10.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/28/2017] [Accepted: 10/18/2017] [Indexed: 12/24/2022]
Abstract
Vitamin A deficiency (A-) is a worldwide public health problem. To better understand how vitamin A status influences gut microbiota and host metabolism, we systematically analyzed urine, cecum, serum and liver samples from vitamin A sufficient (A+) and deficient (A-) mice using 1H NMR-based metabolomics, quantitative (q)PCR and 16S rRNA gene sequencing coupled with multivariate data analysis. The microbiota in the cecum of A- mice showed compositional as well as functional shifts compared to the microbiota from A+ mice. Targeted 1H NMR analyses revealed significant changes in microbial metabolite concentrations including higher butyrate and hippurate and decreased acetate and 4-hydroxyphenylacetate in A+ relative to A- mice. Bacterial butyrate-producing genes including butyryl-CoA:acetate CoA-transferase and butyrate kinase were significantly higher in bacteria from A+ versus bacteria from A- mice. A- mice had disturbances in multiple metabolic pathways including alterations in energy (hyperglycemia, glycogenesis, TCA cycle and lipoprotein biosynthesis), amino acid and nucleic acid metabolism. A- mice had hyperglycemia, liver dysfunction, changes in bacterial metabolism and altered gut microbial communities. Moreover, integrative analyses indicated a strong correlation between gut microbiota and host energy metabolism pathways in the liver. Vitamin A regulates host and bacterial metabolism, and the result includes alterations in energy homeostasis.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States; CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Robert G Nichols
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States
| | - Jingwei Cai
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States
| | - Margherita T Cantorna
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States.
| |
Collapse
|
82
|
Jia L, Shan K, Pan LL, Feng N, Lv Z, Sun Y, Li J, Wu C, Zhang H, Chen W, Diana J, Sun J, Chen YQ. Clostridium butyricum CGMCC0313.1 Protects against Autoimmune Diabetes by Modulating Intestinal Immune Homeostasis and Inducing Pancreatic Regulatory T Cells. Front Immunol 2017; 8:1345. [PMID: 29097999 PMCID: PMC5654235 DOI: 10.3389/fimmu.2017.01345] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
Recent evidence indicates that indigenous Clostridium species induce colonic regulatory T cells (Tregs), and gut lymphocytes are able to migrate to pancreatic islets in an inflammatory environment. Thus, we speculate that supplementation with the well-characterized probiotics Clostridium butyricum CGMCC0313.1 (CB0313.1) may induce pancreatic Tregs and consequently inhibit the diabetes incidence in non-obese diabetic (NOD) mice. CB0313.1 was administered daily to female NOD mice from 3 to 45 weeks of age. The control group received an equal volume of sterile water. Fasting glucose was measured twice a week. Pyrosequencing of the gut microbiota and flow cytometry of mesenteric lymph node (MLN), pancreatic lymph node (PLN), pancreatic and splenic immune cells were performed to investigate the effect of CB0313.1 treatment. Early oral administration of CB0313.1 mitigated insulitis, delayed the onset of diabetes, and improved energy metabolic dysfunction. Protection may involve increased Tregs, rebalanced Th1/Th2/Th17 cells and changes to a less proinflammatory immunological milieu in the gut, PLN, and pancreas. An increase of α4β7+ (the gut homing receptor) Tregs in the PLN suggests that the mechanism may involve increased migration of gut-primed Tregs to the pancreas. Furthermore, 16S rRNA gene sequencing revealed that CB0313.1 enhanced the Firmicutes/Bacteroidetes ratio, enriched Clostridium-subgroups and butyrate-producing bacteria subgroups. Our results provide the basis for future clinical investigations in preventing type 1 diabetes by oral CB0313.1 administration.
Collapse
Affiliation(s)
- Lingling Jia
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Kai Shan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Wuxi No. 2 Hospital, Wuxi, China
| | - Zhuwu Lv
- Department of Obstetrics, Nanjing Medical University Affiliated Wuxi Renmin Hospital, Wuxi, China
| | - Yajun Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
- Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China
| | - Julien Diana
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institute Necker-Enfants Malades (INEM), Centre National de la Recherche Scienctifique, Unité 8253, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jia Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Yong Q. Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
83
|
Lanis JM, Kao DJ, Alexeev EE, Colgan SP. Tissue metabolism and the inflammatory bowel diseases. J Mol Med (Berl) 2017; 95:905-913. [PMID: 28528514 PMCID: PMC5696119 DOI: 10.1007/s00109-017-1544-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/28/2017] [Accepted: 05/04/2017] [Indexed: 12/19/2022]
Abstract
The intestinal mucosa provides a selective barrier between the anaerobic lumen and a highly metabolic lamina propria. A number of recent studies indicate that acute inflammation of the mucosa can result in tissue hypoxia and associated shifts in tissue metabolism. The activation of hypoxia-inducible factor (HIF) under these conditions has been demonstrated to function as an endogenous molecular cue to promote resolution of inflammation, particularly through the orchestration of barrier repair toward homeostasis. Given the central role of oxygen in tissue metabolism, ongoing studies have defined metabolic endpoints of HIF stabilization as important biomarkers of disease activity. Such findings make HIF and HIF-associated metabolic pathways particularly attractive therapeutic targets in inflammatory bowel disease (IBD). Here, we review the recent literature related to tissue metabolism in IBD.
Collapse
Affiliation(s)
- Jordi M Lanis
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado Denver, RC2 Room 10450, 12700 E. 19th Ave, Aurora, CO, 80045, USA
| | - Daniel J Kao
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado Denver, RC2 Room 10450, 12700 E. 19th Ave, Aurora, CO, 80045, USA
| | - Erica E Alexeev
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado Denver, RC2 Room 10450, 12700 E. 19th Ave, Aurora, CO, 80045, USA
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado Denver, RC2 Room 10450, 12700 E. 19th Ave, Aurora, CO, 80045, USA.
| |
Collapse
|
84
|
Jia L, Li D, Feng N, Shamoon M, Sun Z, Ding L, Zhang H, Chen W, Sun J, Chen YQ. Anti-diabetic Effects of Clostridium butyricum CGMCC0313.1 through Promoting the Growth of Gut Butyrate-producing Bacteria in Type 2 Diabetic Mice. Sci Rep 2017; 7:7046. [PMID: 28765642 PMCID: PMC5539151 DOI: 10.1038/s41598-017-07335-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022] Open
Abstract
Patients with type 2 diabetes (T2D) have decreased butyrate-producing bacteria. We hypothesized that supplementation with butyrate-producing bacteria may exert beneficial effects on T2D. The current study investigated the effects of well-characterized butyrate-producing bacteria Clostridium butyricum CGMCC0313.1 (CB0313.1) on hyperglycemia and associated metabolic dysfunction in two diabetic mouse models. CB0313.1 was administered daily by oral gavage to leptindb/db mice for 5 weeks starting from 3 weeks of age, and to HF diabetic mice induced by high fat diet (HFD) plus streptozotocin (STZ) in C57BL/6J mice for 13 weeks starting from 4 weeks of age. CB0313.1 improved diabetic markers (fasting glucose, glucose tolerance, insulin tolerance, GLP-1 and insulin secretion), and decreased blood lipids and inflammatory tone. Furthermore, CB0313.1 reversed hypohepatias and reduced glucose output. We also found that CB0313.1 modulated gut microbiota composition, characterized by a decreased ratio of Firmicutes to Bacteroidetes, reduced Allobaculum bacteria that were abundant in HF diabetic mice and increased butyrate-producing bacteria. Changes in gut microbiota following CB0313.1 treatment were associated with enhanced peroxisome proliferator–activated receptor-γ (PPARγ), insulin signaling molecules and mitochondrial function markers. Together, our study suggests that CB0313.1 may act as a beneficial probiotic for the prevention and treatment of hyperglycemia and associated metabolic dysfunction.
Collapse
Affiliation(s)
- Lingling Jia
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, P. R. China
| | - Dongyao Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, P. R. China.,Wuxi No. 2 Hospital, Jiangsu, P. R. China
| | - Muhammad Shamoon
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Zhenghua Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Lei Ding
- Department of Biology and Chemistry, University Bremen. Leobener Str., NW 2, 28359, Bremen, Germany
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China. .,Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, P. R. China.
| | - Yong Q Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China. .,Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, P. R. China. .,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
85
|
Liu XF, Wei ZY, Bai CL, Ding XB, Li X, Su GH, Cheng L, Zhang L, Guo H, Li GP. Insights into the function of n-3 PUFAs in fat-1 transgenic cattle. J Lipid Res 2017. [PMID: 28626062 DOI: 10.1194/jlr.m072983] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The n-3 PUFAs have many beneficial effects on human health, including roles in immunity, neurodevelopment, and preventing cardiovascular disease. In this study, we established reliable model fat-1 transgenic cattle using transgenic technology and performed a systematic investigation to examine the function of n-3 PUFAs. Our results showed that expression of the fat-1 gene improved several biochemical parameters related to liver function and to plasma glucose and plasma lipid metabolism. Results of global gene and plasma protein expression analysis showed that 310 genes and 13 plasma proteins differed significantly in the blood of fat-1 transgenic cattle compared with WT cattle, reflecting their regulatory roles in the immune and cardiovascular systems. Finally, changes in the gut microflora were also noted in the fat-1 transgenic cattle, suggesting novel roles for n-3 PUFAs in the metabolism of glucose and lipids, as well as anti-stress properties. To the best of our knowledge, this is the first report using multiple parallel analyses to investigate the role of n-3 PUFAs using models such as fat-1 transgenic cattle. This study provides novel insights into the regulatory mechanism of fat-1 in the immune and cardiovascular systems, as well as its anti-stress role.
Collapse
Affiliation(s)
- Xin-Feng Liu
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China; College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin 300384, China
| | - Zhu-Ying Wei
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China
| | - Chun-Ling Bai
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China
| | - Xiang-Bin Ding
- College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin 300384, China
| | - Xin Li
- College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin 300384, China
| | - Guang-Hua Su
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China
| | - Lei Cheng
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China
| | - Li Zhang
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China
| | - Hong Guo
- College of Animal Science and Animal Medicine, Tianjin Agriculture University, Tianjin 300384, China.
| | - Guang-Peng Li
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot 010070, China.
| |
Collapse
|
86
|
Šket R, Treichel N, Debevec T, Eiken O, Mekjavic I, Schloter M, Vital M, Chandler J, Tiedje JM, Murovec B, Prevoršek Z, Stres B. Hypoxia and Inactivity Related Physiological Changes (Constipation, Inflammation) Are Not Reflected at the Level of Gut Metabolites and Butyrate Producing Microbial Community: The PlanHab Study. Front Physiol 2017; 8:250. [PMID: 28522975 PMCID: PMC5416748 DOI: 10.3389/fphys.2017.00250] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/10/2017] [Indexed: 12/12/2022] Open
Abstract
We explored the assembly of intestinal microbiota in healthy male participants during the run-in (5 day) and experimental phases [21-day normoxic bed rest (NBR), hypoxic bedrest (HBR)], and hypoxic ambulation (HAmb) in a strictly controlled laboratory environment, balanced fluid, and dietary intakes, controlled circadian rhythm, microbial ambiental burden, and 24/7 medical surveillance. The fraction of inspired O2 (FiO2) and partial pressure of inspired O2 (PiO2) were 0.209 and 133.1 ± 0.3 mmHg for NBR and 0.141 ± 0.004 and 90.0 ± 0.4 mmHg for both hypoxic variants (HBR and HAmb; ~4,000 m simulated altitude), respectively. A number of parameters linked to intestinal transit spanning Bristol Stool Scale, defecation rates, zonulin, α1-antitrypsin, eosinophil derived neurotoxin, bile acids, reducing sugars, short chain fatty acids, total soluble organic carbon, water content, diet composition, and food intake were measured (167 variables). The abundance, structure, and diversity of butyrate producing microbial community were assessed using the two primary bacterial butyrate synthesis pathways, butyryl-CoA: acetate CoA-transferase (but) and butyrate kinase (buk) genes. Inactivity negatively affected fecal consistency and in combination with hypoxia aggravated the state of gut inflammation (p < 0.05). In contrast, gut permeability, various metabolic markers, the structure, diversity, and abundance of butyrate producing microbial community were not significantly affected. Rearrangements in the butyrate producing microbial community structure were explained by experimental setup (13.4%), experimentally structured metabolites (12.8%), and gut metabolite-immunological markers (11.9%), with 61.9% remaining unexplained. Many of the measured parameters were found to be correlated and were hence omitted from further analyses. The observed progressive increase in two immunological intestinal markers suggested that the transition from healthy physiological state toward the developed symptoms of low magnitude obesity-related syndromes was primarily driven by the onset of inactivity (lack of exercise in NBR) that were exacerbated by systemic hypoxia (HBR) and significantly alleviated by exercise, despite hypoxia (HAmb). Butyrate producing community in colon exhibited apparent resilience toward short-term modifications in host exercise or hypoxia. Progressive constipation (decreased intestinal motility) and increased local inflammation marker suggest that changes in microbial colonization and metabolism were taking place at the location of small intestine.
Collapse
Affiliation(s)
- Robert Šket
- Department of Animal Science, Biotechnical Faculty, University of LjubljanaLjubljana, Slovenia
| | - Nicole Treichel
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München - German Research Center for Environmental HealthNeuherberg, Germany
| | - Tadej Debevec
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan InstituteLjubljana, Slovenia
| | - Ola Eiken
- Department of Environmental Physiology, Swedish Aerospace Physiology Centre, Royal Institute of TechnologyStockholm, Sweden
| | - Igor Mekjavic
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan InstituteLjubljana, Slovenia
| | - Michael Schloter
- Research Unit for Comparative Microbiome Analysis, Helmholtz Zentrum München - German Research Center for Environmental HealthNeuherberg, Germany
| | - Marius Vital
- Center for Microbial Ecology, Michigan State UniversityEast Lansing, MI, USA
| | - Jenna Chandler
- Center for Microbial Ecology, Michigan State UniversityEast Lansing, MI, USA
| | - James M Tiedje
- Center for Microbial Ecology, Michigan State UniversityEast Lansing, MI, USA
| | - Boštjan Murovec
- Laboratory for Artificial Sight and Automation, Faculty of Electrical Sciences, University of LjubljanaLjubljana, Slovenia
| | - Zala Prevoršek
- Department of Animal Science, Biotechnical Faculty, University of LjubljanaLjubljana, Slovenia
| | - Blaž Stres
- Department of Animal Science, Biotechnical Faculty, University of LjubljanaLjubljana, Slovenia.,Center for Clinical Neurophysiology, Faculty of Medicine, University of LjubljanaLjubljana, Slovenia
| |
Collapse
|
87
|
Wang Y, Shou JW, Li XY, Zhao ZX, Fu J, He CY, Feng R, Ma C, Wen BY, Guo F, Yang XY, Han YX, Wang LL, Tong Q, You XF, Lin Y, Kong WJ, Si SY, Jiang JD. Berberine-induced bioactive metabolites of the gut microbiota improve energy metabolism. Metabolism 2017; 70:72-84. [PMID: 28403947 DOI: 10.1016/j.metabol.2017.02.003] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/02/2017] [Accepted: 02/03/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Berberine (BBR) clinically lowers blood lipid and glucose levels via multi-target mechanisms. One of the possible mechanisms is related to its effect on the short chain fatty acids (SCFAs) of the gut microbiota. The goal of this study is to investigate the therapeutic effect and mode of action of BBR working through SCFAs of the gut microbiota (especially, butyrate). METHODS Gas chromatography (GC) was used to detect butyrate and other SCFAs chemically. The effect of BBR on butyrate production was investigated in vitro as well as in several animal systems. Microarrays were used to analyze the composition change in the intestinal bacteria community after treatment with BBR. BBR-induced change in the energy production and gene regulation of intestinal bacteria was examined in order to elucidate the underlying molecular mechanisms. RESULTS We show that oral administration of BBR in animals promoted the gut microbiota to produce butyrate, which then enters the blood and reduces blood lipid and glucose levels. Incubating gut bacterial strains in vitro with BBR increased butyrate production. Orally treating animals directly with butyrate reduced blood lipid and glucose levels through a mechanism different from that of BBR. Intraperitoneal BBR administration did not increase butyrate but reduced blood lipid and glucose levels, suggesting that BBR has two modes of action: the direct effect of the circulated BBR and the indirect effect working through butyrate of the gut microbiota. Pre-treating animals orally with antibiotics abolished the effect of BBR on butyrate. A mechanism study showed that BBR (given orally) modified mice intestinal bacterial composition by increasing the abundance of butyrate-producing bacteria. Furthermore, BBR suppressed bacterial ATP production and NADH levels, resulting in increased butyryl-CoA and, eventually, butyrate production via upregulating phosphotransbutyrylase/butyrate kinase and butyryl-CoA:acetate-CoA transferase in bacteria. CONCLUSION Promotion of butyrate (etc) production in gut microbiota might be one of the important mechanisms of BBR in regulating energy metabolism.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China.
| | - Jia-Wen Shou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Xiao-Yang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Zhen-Xiong Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Jie Fu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Chi-Yu He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Ru Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Chao Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Bao-Ying Wen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Fang Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Xin-Yi Yang
- Insitute of Medicinal Biotechnology, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Yan-Xing Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Lu-Lu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Qian Tong
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Xue-Fu You
- Insitute of Medicinal Biotechnology, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Yuan Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Wei-Jia Kong
- Insitute of Medicinal Biotechnology, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Shu-Yi Si
- Insitute of Medicinal Biotechnology, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China
| | - Jian-Dong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China; Insitute of Medicinal Biotechnology, Chinese Academy of Medical Sciences / Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
88
|
Chun C, Zheng L, Colgan SP. Tissue metabolism and host-microbial interactions in the intestinal mucosa. Free Radic Biol Med 2017; 105:86-92. [PMID: 27687211 PMCID: PMC5797701 DOI: 10.1016/j.freeradbiomed.2016.09.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 01/03/2023]
Abstract
In recent years, studies in the gastrointestinal (GI) mucosa have taught us a number of important lessons related to tissue oxygenation and metabolism in health and disease. The highly vascularized mucosa lies immediately adjacent to an anaerobic lumen containing trillions of metabolically active microbes (i.e. the microbiome) that results in one of the more austere tissue microenvironments in the body. These studies have also implicated a prominent role for oxygen metabolism and hypoxia in inflammation, so called "inflammatory hypoxia", that results from the activation of multiple oxygen consuming enzymes. Inflammation-associated shifts in the composition of the microbiome and microbial-derived metabolites have revealed a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of key target genes that promote inflammatory resolution. Analyses of these pathways have provided a multitude of opportunities for understanding basic mechanisms of both homeostasis and disease and have defined new targets for intervention. Here, we review recent advances in our understanding of metabolic influences on host-microbe interactions in the GI mucosa.
Collapse
Affiliation(s)
- Carlene Chun
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
| | - Leon Zheng
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|
89
|
EGCG Prevents High Fat Diet-Induced Changes in Gut Microbiota, Decreases of DNA Strand Breaks, and Changes in Expression and DNA Methylation of Dnmt1 and MLH1 in C57BL/6J Male Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3079148. [PMID: 28133504 PMCID: PMC5241499 DOI: 10.1155/2017/3079148] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/12/2016] [Accepted: 10/20/2016] [Indexed: 12/11/2022]
Abstract
Obesity as a multifactorial disorder involves low-grade inflammation, increased reactive oxygen species incidence, gut microbiota aberrations, and epigenetic consequences. Thus, prevention and therapies with epigenetic active antioxidants, (-)-Epigallocatechin-3-gallate (EGCG), are of increasing interest. DNA damage, DNA methylation and gene expression of DNA methyltransferase 1, interleukin 6, and MutL homologue 1 were analyzed in C57BL/6J male mice fed a high-fat diet (HFD) or a control diet (CD) with and without EGCG supplementation. Gut microbiota was analyzed with quantitative real-time polymerase chain reaction. An induction of DNA damage was observed, as a consequence of HFD-feeding, whereas EGCG supplementation decreased DNA damage. HFD-feeding induced a higher inflammatory status. Supplementation reversed these effects, resulting in tissue specific gene expression and methylation patterns of DNA methyltransferase 1 and MutL homologue 1. HFD feeding caused a significant lower bacterial abundance. The Firmicutes/Bacteroidetes ratio is significantly lower in HFD + EGCG but higher in CD + EGCG compared to control groups. The results demonstrate the impact of EGCG on the one hand on gut microbiota which together with dietary components affects host health. On the other hand effects may derive from antioxidative activities as well as epigenetic modifications observed on CpG methylation but also likely to include other epigenetic elements.
Collapse
|
90
|
Machiels K, Sabino J, Vandermosten L, Joossens M, Arijs I, de Bruyn M, Eeckhaut V, Van Assche G, Ferrante M, Verhaegen J, Van Steen K, Van Immerseel F, Huys G, Verbeke K, Wolthuis A, de Buck Van Overstraeten A, D'Hoore A, Rutgeerts P, Vermeire S. Specific members of the predominant gut microbiota predict pouchitis following colectomy and IPAA in UC. Gut 2017; 66:79-88. [PMID: 26423113 DOI: 10.1136/gutjnl-2015-309398] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Pouchitis is the most common complication after colectomy with ileal pouch-anal anastomosis (IPAA) for UC and the risk is the highest within the 1st year after surgery. The pathogenesis is not completely understood but clinical response to antibiotics suggests a role for gut microbiota. We hypothesised that the risk for pouchitis can be predicted based on the faecal microbial composition before colectomy. DESIGN Faecal samples from 21 patients with UC undergoing IPAA were prospectively collected before colectomy and at predefined clinical visits at 1 month, 3 months, 6 months and 12 months after IPAA. The predominant microbiota was analysed using community profiling with denaturing gradient gel electrophoresis followed by quantitative real-time PCR validation. RESULTS Cluster analysis before colectomy distinguished patients with pouchitis from those with normal pouch during the 1st year of follow-up. In patients developing pouchitis, an increase of Ruminococcus gnavus (p<0.001), Bacteroides vulgatus (p=0.043), Clostridium perfringens (p=0.011) and a reduction of two Lachnospiraceae genera (Blautia (p=0.04), Roseburia (p=0.008)) was observed. A score combining these five bacterial risk factors was calculated and presence of at least two risk factors showed a sensitivity and specificity of 100% and 63.6%, respectively. CONCLUSIONS Presence of R. gnavus, B. vulgatus and C. perfringens and absence of Blautia and Roseburia in faecal samples of patients with UC before surgery is associated with a higher risk of pouchitis after IPAA. Our findings suggest new predictive and therapeutic strategies in patients undergoing colectomy with IPAA.
Collapse
Affiliation(s)
- Kathleen Machiels
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - João Sabino
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Leen Vandermosten
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Marie Joossens
- Department Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Center for the Biology of Disease, VIB, Leuven, Belgium.,Faculty of Sciences and Bioengineering Sciences, Microbiology Unit, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ingrid Arijs
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Magali de Bruyn
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Venessa Eeckhaut
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Merelbeke, Belgium
| | - Gert Van Assche
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Marc Ferrante
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Jan Verhaegen
- Department of Microbiology and Immunology, University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Kristel Van Steen
- Department of Electrical Engineering and Computer Science, Montefiore Institute, Liège, Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Merelbeke, Belgium
| | - Geert Huys
- Laboratory of Microbiology & BCCM/LMG Bacteria Collection, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Albert Wolthuis
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | | | - Andre D'Hoore
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Paul Rutgeerts
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| |
Collapse
|
91
|
Miyoshi J, Chang EB. The gut microbiota and inflammatory bowel diseases. Transl Res 2017; 179:38-48. [PMID: 27371886 PMCID: PMC5156589 DOI: 10.1016/j.trsl.2016.06.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/02/2016] [Accepted: 06/06/2016] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel diseases (IBDs) are chronic diseases of unclear etiology that affect over 1 million individuals in the United States and over 2.5 million people in Europe. However, they are also expanding globally, affecting populations in Asia, South America, and the Middle East as they become more industrialized. These diseases are believed to arise from the convergence of genetic, environmental, and microbial factors that trigger aberrant immune and tissue responses, resulting in intestinal inflammation. Advances in cultivation-independent investigations, experimental models, and bioinformatics approaches have improved our understanding of the role of gut microbiota in IBD. However, determining and understanding the functional consequences of gut dysbiosis and altered host-microbiota interactions in IBD remain a challenge due to the limits of current experimental models and difficulty in establishing causal links in human-based investigations. Continued development of new methodologies and improvements in clinical study design are needed to better understand the interplay of genetic, microbial, and immunological factors in IBD. This knowledge can then be applied clinically to improve therapeutic strategies and outcomes for IBD.
Collapse
Affiliation(s)
- Jun Miyoshi
- Department of Medicine, The University of Chicago, Chicago, Ill
| | - Eugene B Chang
- Department of Medicine, The University of Chicago, Chicago, Ill.
| |
Collapse
|
92
|
Anand S, Kaur H, Mande SS. Comparative In silico Analysis of Butyrate Production Pathways in Gut Commensals and Pathogens. Front Microbiol 2016; 7:1945. [PMID: 27994578 PMCID: PMC5133246 DOI: 10.3389/fmicb.2016.01945] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/18/2016] [Indexed: 12/22/2022] Open
Abstract
Biosynthesis of butyrate by commensal bacteria plays a crucial role in maintenance of human gut health while dysbiosis in gut microbiome has been linked to several enteric disorders. Contrastingly, butyrate shows cytotoxic effects in patients with oral diseases like periodontal infections and oral cancer. In addition to these host associations, few syntrophic bacteria couple butyrate degradation with sulfate reduction and methane production. Thus, it becomes imperative to understand the distribution of butyrate metabolism pathways and delineate differences in substrate utilization between pathogens and commensals. The bacteria utilize four pathways for butyrate production with different initial substrates (Pyruvate, 4-aminobutyrate, Glutarate and Lysine) which follow a polyphyletic distribution. A comprehensive mining of complete/draft bacterial genomes indicated conserved juxtaposed genomic arrangement in all these pathways. This gene context information was utilized for an accurate annotation of butyrate production pathways in bacterial genomes. Interestingly, our analysis showed that inspite of a beneficial impact of butyrate in gut, not only commensals, but a few gut pathogens also possess butyrogenic pathways. The results further illustrated that all the gut commensal bacteria (Faecalibacterium, Roseburia, Butyrivibrio, and commensal species of Clostridia etc) ferment pyruvate for butyrate production. On the contrary, the butyrogenic gut pathogen Fusobacterium utilizes different amino acid metabolism pathways like those for Glutamate (4-aminobutyrate and Glutarate) and Lysine for butyrogenesis which leads to a concomitant release of harmful by-products like ammonia in the process. The findings in this study indicate that commensals and pathogens in gut have divergently evolved to produce butyrate using distinct pathways. No such evolutionary selection was observed in oral pathogens (Porphyromonas and Filifactor) which showed presence of pyruvate as well as amino acid fermenting pathways which might be because the final product butyrate is itself known to be cytotoxic in oral diseases. This differential utilization of butyrogenic pathways in gut pathogens and commensals has an enormous ecological impact taking into consideration the immense influence of butyrate on different disorders in humans. The results of this study can potentially guide bioengineering experiments to design therapeutics/probiotics by manipulation of butyrate biosynthesis gene clusters in bacteria.
Collapse
Affiliation(s)
- Swadha Anand
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd. Pune, India
| | - Harrisham Kaur
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd. Pune, India
| | - Sharmila S Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd. Pune, India
| |
Collapse
|
93
|
Trachsel J, Bayles DO, Looft T, Levine UY, Allen HK. Function and Phylogeny of Bacterial Butyryl Coenzyme A:Acetate Transferases and Their Diversity in the Proximal Colon of Swine. Appl Environ Microbiol 2016; 82:6788-6798. [PMID: 27613689 PMCID: PMC5086572 DOI: 10.1128/aem.02307-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/07/2016] [Indexed: 12/11/2022] Open
Abstract
Studying the host-associated butyrate-producing bacterial community is important, because butyrate is essential for colonic homeostasis and gut health. Previous research has identified the butyryl coenzyme A (CoA):acetate-CoA transferase (EC 2.3.8.3) as a gene of primary importance for butyrate production in intestinal ecosystems; however, this gene family (but) remains poorly defined. We developed tools for the analysis of butyrate-producing bacteria based on 12 putative but genes identified in the genomes of nine butyrate-producing bacteria obtained from the swine intestinal tract. Functional analyses revealed that eight of these genes had strong But enzyme activity. When but paralogues were found within a genome, only one gene per genome encoded strong activity, with the exception of one strain in which no gene encoded strong But activity. Degenerate primers were designed to amplify the functional but genes and were tested by amplifying environmental but sequences from DNA and RNA extracted from swine colonic contents. The results show diverse but sequences from swine-associated butyrate-producing bacteria, most of which clustered near functionally confirmed sequences. Here, we describe tools and a framework that allow the bacterial butyrate-producing community to be profiled in the context of animal health and disease. IMPORTANCE Butyrate is a compound produced by the microbiota in the intestinal tracts of animals. This compound is of critical importance for intestinal health, and yet studying its production by diverse intestinal bacteria is technically challenging. Here, we present an additional way to study the butyrate-producing community of bacteria using one degenerate primer set that selectively targets genes experimentally demonstrated to encode butyrate production. This work will enable researchers to more easily study this very important bacterial function that has implications for host health and resistance to disease.
Collapse
Affiliation(s)
- Julian Trachsel
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, USA Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa, USA
| | - Darrell O Bayles
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, USA
| | - Torey Looft
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, USA
| | - Uri Y Levine
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, USA
| | - Heather K Allen
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, USA
| |
Collapse
|
94
|
Sato H, Zhang LS, Martinez K, Chang EB, Yang Q, Wang F, Howles PN, Hokari R, Miura S, Tso P. Antibiotics Suppress Activation of Intestinal Mucosal Mast Cells and Reduce Dietary Lipid Absorption in Sprague-Dawley Rats. Gastroenterology 2016; 151:923-932. [PMID: 27436071 PMCID: PMC5391873 DOI: 10.1053/j.gastro.2016.07.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/28/2016] [Accepted: 07/08/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.
Collapse
Affiliation(s)
- Hirokazu Sato
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Linda S Zhang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kristina Martinez
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Qing Yang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Fei Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Philip N Howles
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Soichiro Miura
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
95
|
Dehoux P, Marvaud JC, Abouelleil A, Earl AM, Lambert T, Dauga C. Comparative genomics of Clostridium bolteae and Clostridium clostridioforme reveals species-specific genomic properties and numerous putative antibiotic resistance determinants. BMC Genomics 2016; 17:819. [PMID: 27769168 PMCID: PMC5073890 DOI: 10.1186/s12864-016-3152-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Clostridium bolteae and Clostridium clostridioforme, previously included in the complex C. clostridioforme in the group Clostridium XIVa, remain difficult to distinguish by phenotypic methods. These bacteria, prevailing in the human intestinal microbiota, are opportunistic pathogens with various drug susceptibility patterns. In order to better characterize the two species and to obtain information on their antibiotic resistance genes, we analyzed the genomes of six strains of C. bolteae and six strains of C. clostridioforme, isolated from human infection. RESULTS The genome length of C. bolteae varied from 6159 to 6398 kb, and 5719 to 6059 CDSs were detected. The genomes of C. clostridioforme were smaller, between 5467 and 5927 kb, and contained 5231 to 5916 CDSs. The two species display different metabolic pathways. The genomes of C. bolteae contained lactose operons involving PTS system and complex regulation, which contribute to phenotypic differentiation from C. clostridioforme. The Acetyl-CoA pathway, similar to that of Faecalibacterium prausnitzii, a major butyrate producer in the human gut, was only found in C. clostridioforme. The two species have also developed diverse flagella mobility systems contributing to gut colonization. Their genomes harboured many CDSs involved in resistance to beta-lactams, glycopeptides, macrolides, chloramphenicol, lincosamides, rifampin, linezolid, bacitracin, aminoglycosides and tetracyclines. Overall antimicrobial resistance genes were similar within a species, but strain-specific resistance genes were found. We discovered a new group of genes coding for rifampin resistance in C. bolteae. C. bolteae 90B3 was resistant to phenicols and linezolide in producing a 23S rRNA methyltransferase. C. clostridioforme 90A8 contained the VanB-type Tn1549 operon conferring vancomycin resistance. We also detected numerous genes encoding proteins related to efflux pump systems. CONCLUSION Genomic comparison of C. bolteae and C. clostridiofrome revealed functional differences in butyrate pathways and in flagellar systems, which play a critical role within human microbiota. Most of the resistance genes detected in both species were previously characterized in other bacterial species. A few of them were related to antibiotics inactive against Clostridium spp. Some were part of mobile genetic elements suggesting that these commensals of the human microbiota act as reservoir of antimicrobial resistances.
Collapse
Affiliation(s)
- Pierre Dehoux
- Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - Jean Christophe Marvaud
- Faculté de Pharmacie, EA4043 “Unité Bactéries Pathogènes et Santé” (UBaPS), Université Paris Sud, Châtenay-Malabry Cedex, 92296 France
| | - Amr Abouelleil
- Genome Sequencing and Analysis Program, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Ashlee M. Earl
- Genome Sequencing and Analysis Program, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Thierry Lambert
- Faculté de Pharmacie, EA4043 “Unité Bactéries Pathogènes et Santé” (UBaPS), Université Paris Sud, Châtenay-Malabry Cedex, 92296 France
- Antibacterial Agents Unit, Department of Microbiology, Institut Pasteur, Paris, France
| | - Catherine Dauga
- Department of Genomes and Genetics, Institut Pasteur, Paris, France
- International Group of Data Analysis, Centre for Bioinformatics, Biostatistics and Integrative Biology, Institut Pasteur, Paris, France
| |
Collapse
|
96
|
Meisel M, Mayassi T, Fehlner-Peach H, Koval JC, O'Brien SL, Hinterleitner R, Lesko K, Kim S, Bouziat R, Chen L, Weber CR, Mazmanian SK, Jabri B, Antonopoulos DA. Interleukin-15 promotes intestinal dysbiosis with butyrate deficiency associated with increased susceptibility to colitis. ISME JOURNAL 2016; 11:15-30. [PMID: 27648810 DOI: 10.1038/ismej.2016.114] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 06/17/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023]
Abstract
Dysbiosis resulting in gut-microbiome alterations with reduced butyrate production are thought to disrupt intestinal immune homeostasis and promote complex immune disorders. However, whether and how dysbiosis develops before the onset of overt pathology remains poorly defined. Interleukin-15 (IL-15) is upregulated in distressed tissue and its overexpression is thought to predispose susceptible individuals to and have a role in the pathogenesis of celiac disease and inflammatory bowel disease (IBD). Although the immunological roles of IL-15 have been largely studied, its potential impact on the microbiota remains unexplored. Analysis of 16S ribosomal RNA-based inventories of bacterial communities in mice overexpressing IL-15 in the intestinal epithelium (villin-IL-15 transgenic (v-IL-15tg) mice) shows distinct changes in the composition of the intestinal bacteria. Although some alterations are specific to individual intestinal compartments, others are found across the ileum, cecum and feces. In particular, IL-15 overexpression restructures the composition of the microbiota with a decrease in butyrate-producing bacteria that is associated with a reduction in luminal butyrate levels across all intestinal compartments. Fecal microbiota transplant experiments of wild-type and v-IL-15tg microbiota into germ-free mice further indicate that diminishing butyrate concentration observed in the intestinal lumen of v-IL-15tg mice is the result of intrinsic alterations in the microbiota induced by IL-15. This reconfiguration of the microbiota is associated with increased susceptibility to dextran sodium sulfate-induced colitis. Altogether, this study reveals that IL-15 impacts butyrate-producing bacteria and lowers butyrate levels in the absence of overt pathology, which represent events that precede and promote intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Marlies Meisel
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Toufic Mayassi
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Jason C Koval
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Sarah L O'Brien
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | | | - Kathryn Lesko
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Sangman Kim
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Romain Bouziat
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Li Chen
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Bana Jabri
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Pathology, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Dionysios A Antonopoulos
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Biosciences Division, Argonne National Laboratory, Argonne, IL, USA.,Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
97
|
Boets E, Gomand SV, Deroover L, Preston T, Vermeulen K, De Preter V, Hamer HM, Van den Mooter G, De Vuyst L, Courtin CM, Annaert P, Delcour JA, Verbeke KA. Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: a stable isotope study. J Physiol 2016; 595:541-555. [PMID: 27510655 DOI: 10.1113/jp272613] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS The short-chain fatty acids (SCFAs) are bacterial metabolites produced during the colonic fermentation of undigested carbohydrates, such as dietary fibre and prebiotics, and can mediate the interaction between the diet, the microbiota and the host. We quantified the fraction of colonic administered SCFAs that could be recovered in the systemic circulation, the fraction that was excreted via the breath and urine, and the fraction that was used as a precursor for glucose, cholesterol and fatty acids. This information is essential for understanding the molecular mechanisms by which SCFAs beneficially affect physiological functions such as glucose and lipid metabolism and immune function. ABSTRACT The short-chain fatty acids (SCFAs), acetate, propionate and butyrate, are bacterial metabolites that mediate the interaction between the diet, the microbiota and the host. In the present study, the systemic availability of SCFAs and their incorporation into biologically relevant molecules was quantified. Known amounts of 13 C-labelled acetate, propionate and butyrate were introduced in the colon of 12 healthy subjects using colon delivery capsules and plasma levels of 13 C-SCFAs 13 C-glucose, 13 C-cholesterol and 13 C-fatty acids were measured. The butyrate-producing capacity of the intestinal microbiota was also quantified. Systemic availability of colonic-administered acetate, propionate and butyrate was 36%, 9% and 2%, respectively. Conversion of acetate into butyrate (24%) was the most prevalent interconversion by the colonic microbiota and was not related to the butyrate-producing capacity in the faecal samples. Less than 1% of administered acetate was incorporated into cholesterol and <15% in fatty acids. On average, 6% of colonic propionate was incorporated into glucose. The SCFAs were mainly excreted via the lungs after oxidation to 13 CO2 , whereas less than 0.05% of the SCFAs were excreted into urine. These results will allow future evaluation and quantification of SCFA production from 13 C-labelled fibres in the human colon by measurement of 13 C-labelled SCFA concentrations in blood.
Collapse
Affiliation(s)
- Eef Boets
- Translational Research in Gastrointestinal Disorders.,Leuven Food Science and Nutrition Research Centre
| | - Sara V Gomand
- Leuven Food Science and Nutrition Research Centre.,Center for Food and Microbial Technology
| | - Lise Deroover
- Translational Research in Gastrointestinal Disorders.,Leuven Food Science and Nutrition Research Centre
| | - Tom Preston
- Stable Isotope Biochemistry Laboratory, Scottish Universities Environmental Research Centre, University of Glasgow, Glasgow, UK
| | - Karen Vermeulen
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Merelbeke, Belgium
| | - Vicky De Preter
- Translational Research in Gastrointestinal Disorders.,Group Health and Social Work, UC Leuven-Limburg, Leuven, Belgium
| | - Henrike M Hamer
- Translational Research in Gastrointestinal Disorders.,Leuven Food Science and Nutrition Research Centre
| | | | - Luc De Vuyst
- Industrial Microbiology and Food Biotechnology, Vrije Universiteit Brussel, Brussel, Belgium
| | - Christophe M Courtin
- Leuven Food Science and Nutrition Research Centre.,Center for Food and Microbial Technology
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Jan A Delcour
- Leuven Food Science and Nutrition Research Centre.,Center for Food and Microbial Technology
| | - Kristin A Verbeke
- Translational Research in Gastrointestinal Disorders.,Leuven Food Science and Nutrition Research Centre
| |
Collapse
|
98
|
Hippe B, Remely M, Aumueller E, Pointner A, Magnet U, Haslberger A. Faecalibacterium prausnitzii phylotypes in type two diabetic, obese, and lean control subjects. Benef Microbes 2016; 7:511-7. [DOI: 10.3920/bm2015.0075] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Faecalibacterium prausnitzii is one of the main butyrate producers in the healthy human gut. Information on its genetic diversity is lacking, although two genetic phylotypes have been differentiated. In the present study, F. prausnitzii phylotypes were examined in faeces of obese and type two diabetes with similar eating behaviour compared to a lean control group. The purpose of the study was to analyse if an excessive butyrate production induced by different F. prausnitzii phylotypes discriminates between obese developing type two diabetes or not. The faecal samples were analysed for the total abundance of F. prausnitzii 16S rRNA copies, fragment lengths polymorphism, high resolution melt curve analysis (HRM) and the butyryl-CoA:acetate CoA-transferase gene copies and melt curve variances. The diabetic group was found to differ significantly from the lean control group in the results of qPCR, butyryl-CoA:acetyate CoA-transferase gene melt curve, and HRM. F. prausnitzii phylotypes differed in obese with and without developed diabetes type two. Different phylotypes of F. prausnitzii may lead to differences in the inflammatory genesis in the host. F. prausnitzii phylotypes may have an influence on developing type two diabetes and might also act as starting points for prevention and therapy of obesity associated disease.
Collapse
Affiliation(s)
- B. Hippe
- Institute of Nutritional Sciences, Althanstr. 14, UZA 2, 1090 Vienna, Austria
| | - M. Remely
- Institute of Nutritional Sciences, Althanstr. 14, UZA 2, 1090 Vienna, Austria
| | - E. Aumueller
- Institute of Nutritional Sciences, Althanstr. 14, UZA 2, 1090 Vienna, Austria
| | - A. Pointner
- Institute of Nutritional Sciences, Althanstr. 14, UZA 2, 1090 Vienna, Austria
| | - U. Magnet
- Institute of Nutritional Sciences, Althanstr. 14, UZA 2, 1090 Vienna, Austria
| | - A.G. Haslberger
- Institute of Nutritional Sciences, Althanstr. 14, UZA 2, 1090 Vienna, Austria
| |
Collapse
|
99
|
Colon microbiota fermentation of dietary prebiotics towards short-chain fatty acids and their roles as anti-inflammatory and antitumour agents: A review. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.06.032] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
100
|
The Microbiome of Animals: Implications for Conservation Biology. Int J Genomics 2016; 2016:5304028. [PMID: 27195280 PMCID: PMC4852354 DOI: 10.1155/2016/5304028] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023] Open
Abstract
In recent years the human microbiome has become a growing area of research and it is becoming clear that the microbiome of humans plays an important role for human health. Extensive research is now going into cataloging and annotating the functional role of the human microbiome. The ability to explore and describe the microbiome of any species has become possible due to new methods for sequencing. These techniques allow comprehensive surveys of the composition of the microbiome of nonmodel organisms of which relatively little is known. Some attention has been paid to the microbiome of insect species including important vectors of pathogens of human and veterinary importance, agricultural pests, and model species. Together these studies suggest that the microbiome of insects is highly dependent on the environment, species, and populations and affects the fitness of species. These fitness effects can have important implications for the conservation and management of species and populations. Further, these results are important for our understanding of invasion of nonnative species, responses to pathogens, and responses to chemicals and global climate change in the present and future.
Collapse
|