$\require{mediawiki-texvc}$

연합인증

연합인증 가입 기관의 연구자들은 소속기관의 인증정보(ID와 암호)를 이용해 다른 대학, 연구기관, 서비스 공급자의 다양한 온라인 자원과 연구 데이터를 이용할 수 있습니다.

이는 여행자가 자국에서 발행 받은 여권으로 세계 각국을 자유롭게 여행할 수 있는 것과 같습니다.

연합인증으로 이용이 가능한 서비스는 NTIS, DataON, Edison, Kafe, Webinar 등이 있습니다.

한번의 인증절차만으로 연합인증 가입 서비스에 추가 로그인 없이 이용이 가능합니다.

다만, 연합인증을 위해서는 최초 1회만 인증 절차가 필요합니다. (회원이 아닐 경우 회원 가입이 필요합니다.)

연합인증 절차는 다음과 같습니다.

최초이용시에는
ScienceON에 로그인 → 연합인증 서비스 접속 → 로그인 (본인 확인 또는 회원가입) → 서비스 이용

그 이후에는
ScienceON 로그인 → 연합인증 서비스 접속 → 서비스 이용

연합인증을 활용하시면 KISTI가 제공하는 다양한 서비스를 편리하게 이용하실 수 있습니다.

Synbiotic Supplementation Containing Whole Plant Sugar Cane Fibre and Probiotic Spores Potentiates Protective Synergistic Effects in Mouse Model of IBD 원문보기

Nutrients, v.11 no.4, 2019년, pp.818 -   

Shinde, Tanvi ,  Perera, Agampodi Promoda ,  Vemuri, Ravichandra ,  Gondalia, Shakuntla V. ,  Karpe, Avinash V. ,  Beale, David J. ,  Shastri, Sonia ,  Southam, Benjamin ,  Eri, Rajaraman ,  Stanley, Roger

Abstract AI-Helper 아이콘AI-Helper

Inflammatory bowel diseases (IBD) are a chronic inflammatory disorders with increasing global incidence. Synbiotic, which is a two-point approach carrying probiotic and prebiotic components in mitigating inflammation in IBD, is thought to be a pragmatic approach owing to the synergistic outcomes. In...

참고문헌 (81)

  1. Vindigni, Stephen M., Zisman, Timothy L., Suskind, David L., Damman, Christopher J.. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: a tripartite pathophysiological circuit with implications for new therapeutic directions. Therapeutic advances in gastroenterology, vol.9, no.4, 606-625.

  2. Ng, Siew C, Shi, Hai Yun, Hamidi, Nima, Underwood, Fox E, Tang, Whitney, Benchimol, Eric I, Panaccione, Remo, Ghosh, Subrata, Wu, Justin C Y, Chan, Francis K L, Sung, Joseph J Y, Kaplan, Gilaad G. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. The Lancet, vol.390, no.10114, 2769-2778.

  3. Hou, Jason K., Lee, Dale, Lewis, James. Diet and Inflammatory Bowel Disease: Review of Patient-Targeted Recommendations. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association, vol.12, no.10, 1592-1600.

  4. Palumbo, Vincenzo Davide, Romeo, Marcello, Gammazza, Antonella Marino, Carini, Francesco, Damiani, Provvidenza, Damiano, Giuseppe, Buscemi, Salvatore, Lo Monte, Attilio Ignazio, Gerges-Geagea, Alice, Jurjus, Abdo, Tomasello, Giovanni. The long-term effects of probiotics in the therapy of ulcerative colitis: A clinical study. Biomedical papers of the Medical Faculty of the University Palacký, Olomouc, Czech Republic, vol.160, no.3, 372-377.

  5. Abruzzo The influence of some dietary components on intestinal microbiota Prog. Nutr. 2016 18 205 

  6. Steed, H., Macfarlane, G. T., Blackett, K. L., Bahrami, B., Reynolds, N., Walsh, S. V., Cummings, J. H., Macfarlane, S.. Clinical trial: the microbiological and immunological effects of synbiotic consumption - a randomized double‐blind placebo‐controlled study in active Crohn’s disease. Alimentary pharmacology & therapeutics, vol.32, no.7, 872-883.

  7. Wasilewski, Andrzej, Zielińska, Marta, Storr, Martin, Fichna, Jakub. Beneficial Effects of Probiotics, Prebiotics, Synbiotics, and Psychobiotics in Inflammatory Bowel Disease :. Inflammatory bowel diseases, vol.21, no.7, 1674-1682.

  8. Moroeanu, Veronica Ionela, Vamanu, Emanuel, Paun, Gabriela, Neagu, Elena, Ungureanu, Oana Rodica, Eremia, Sandra A. V., Radu, Gabriel-Lucian, Ionescu, Robertina, Pelinescu, Diana Roxana. Probiotic Strains Influence on Infant Microbiota in the In Vitro Colonic Fermentation Model GIS1. Indian journal of microbiology, vol.55, no.4, 423-429.

  9. Wong, Celestine, Harris, Philip J., Ferguson, Lynnette R.. Potential Benefits of Dietary Fibre Intervention in Inflammatory Bowel Disease. International journal of molecular sciences, vol.17, no.6, 919-.

  10. Pituch-Zdanowska, Aleksandra, Banaszkiewicz, Aleksandra, Albrecht, Piotr. The role of dietary fibre in inflammatory bowel disease. Przegląd gastroenterologiczny. = Gastroenterology review, vol.10, no.3, 135-141.

  11. Galvez, Julio, Rodríguez-Cabezas, M. Elena, Zarzuelo, Antonio. Effects of dietary fiber on inflammatory bowel disease. Molecular nutrition & food research, vol.49, no.6, 601-608.

  12. Williams, Barbara A., Grant, Lucas J., Gidley, Michael J., Mikkelsen, Deirdre. Gut Fermentation of Dietary Fibres: Physico-Chemistry of Plant Cell Walls and Implications for Health. International journal of molecular sciences, vol.18, no.10, 2203-.

  13. Tuohy, Kieran M., Conterno, Lorenza, Gasperotti, Mattia, Viola, Roberto. Up-regulating the Human Intestinal Microbiome Using Whole Plant Foods, Polyphenols, and/or Fiber. Journal of agricultural and food chemistry, vol.60, no.36, 8776-8782.

  14. Liu, Rui Hai. Health benefits of fruit and vegetables are from additive and synergistic combinations of phytochemicals. The American journal of clinical nutrition, vol.78, no.3, 517S-520S.

  15. Gamage, Hasinika K. A. H., Tetu, Sasha G., Chong, Raymond W. W., Bucio-Noble, Daniel, Rosewarne, Carly P., Kautto, Liisa, Ball, Malcolm S., Molloy, Mark P., Packer, Nicolle H., Paulsen, Ian T.. Fiber Supplements Derived From Sugarcane Stem, Wheat Dextrin and Psyllium Husk Have Different In Vitro Effects on the Human Gut Microbiota. Frontiers in microbiology, vol.9, 1618-.

  16. Walton, Sara L., Bischoff, Kenneth M., van Heiningen, Adriaan R. P., van Walsum, G. Peter. Production of lactic acid from hemicellulose extracts by Bacillus coagulans MXL-9. Journal of industrial microbiology & biotechnology, vol.37, no.8, 823-830.

  17. Konuray, Gözde, Erginkaya, Zerrin. Potential Use of Bacillus coagulans in the Food Industry. Foods, vol.7, no.6, 92-.

  18. Slavin, Joanne L., Brauer, Paula M., Marlett, Judith A.. Neutral Detergent Fiber, Hemicellulose and Cellulose Digestibility in Human Subjects. The Journal of nutrition, vol.111, no.2, 287-297.

  19. Leschine, S B. Cellulose Degradation in Anaerobic Environments. Annual review of microbiology, vol.49, 399-426.

  20. Shinde, Tanvi, Vemuri, Ravichandra, Shastri, Madhur D., Perera, Agampodi Promoda, Tristram, Stephen, Stanley, Roger, Eri, Rajaraman. Probiotic Bacillus coagulans MTCC 5856 spores exhibit excellent in-vitro functional efficacy in simulated gastric survival, mucosal adhesion and immunomodulation. Journal of functional foods : the official journal of the International Society for Nutraceuticals & Functional Foods, vol.52, 100-108.

  21. Baron, Mira. Original Research: A Patented Strain ofBacillus coagulansIncreased Immune Response to Viral Challenge. Postgraduate medicine, vol.121, no.2, 114-118.

  22. Chassaing Dextran sulfate sodium (dss)-induced colitis in mice Curr. Protoc. Immunol. 2014 10.1002/0471142735.im1525s104 104 15.25. 1 

  23. Murthy, S. N. S., Cooper, Harry S., Shim, Helen, Shah, Rupal S., Ibrahim, Samih A., Sedergran, Deborah J.. Treatment of dextran sulfate sodium-induced murine colitis by intracolonic cyclosporin. Digestive diseases and sciences, vol.38, no.9, 1722-1734.

  24. Perera, Agampodi Promoda, Fernando, Ruchira, Shinde, Tanvi, Gundamaraju, Rohit, Southam, Benjamin, Sohal, Sukhwinder Singh, Robertson, Avril A. B., Schroder, Kate, Kunde, Dale, Eri, Rajaraman. MCC950, a specific small molecule inhibitor of NLRP3 inflammasome attenuates colonic inflammation in spontaneous colitis mice. Scientific reports, vol.8, 8618-8618.

  25. Demon, Dieter, Kuchmiy, Anna, Fossoul, Amelie, Zhu, Qifan, Kanneganti, Thirumala-Devi, Lamkanfi, Mohamed. Caspase-11 is expressed in the colonic mucosa and protects against dextran sodium sulphate-induced colitis. Mucosal immunology, vol.7, no.6, 1480-1491.

  26. Eri T cell transfer model of colitis: A great tool to assess the contribution of t cells in chronic intestinal inflammation Leucocytes 2012 261 

  27. Koelink, Pim J, Wildenberg, Manon E, Stitt, Larry W, Feagan, Brian G, Koldijk, Martin, van ‘t Wout, Angélique B, Atreya, Raja, Vieth, Michael, Brandse, Johannan F, Duijst, Suzanne, te Velde, Anje A, D’Haens, Geert R A M, Levesque, Barrett G, van den Brink, Gijs R. Development of Reliable, Valid and Responsive Scoring Systems for Endoscopy and Histology in Animal Models for Inflammatory Bowel Disease. Journal of Crohn’s and colitis, vol.12, no.7, 794-803.

  28. Sovran, Bruno, Lu, Peng, Loonen, Linda M.P., Hugenholtz, Floor, Belzer, Clara, Stolte, Ellen H., Boekschoten, Mark V., van Baarlen, Peter, Smidt, Hauke, Kleerebezem, Michiel, de Vos, Paul, Renes, Ingrid B., Wells, Jerry M., Dekker, Jan. Identification of Commensal Species Positively Correlated with Early Stress Responses to a Compromised Mucus Barrier :. Inflammatory bowel diseases, vol.22, no.4, 826-840.

  29. Li, Pei, Zhang, Ruijie, Wang, Liyuan, Gan, Yibo, Xu, Yuan, Song, Lei, Luo, Lei, Zhao, Chen, Zhang, Chengmin, Ouyang, Bin, Tu, Bing, Zhou, Qiang. Long-term load duration induces N-cadherin down-regulation and loss of cell phenotype of nucleus pulposus cells in a disc bioreactor culture. Bioscience reports, vol.37, no.2, BSR20160582-.

  30. Lean, Qi Ying, Eri, Rajaraman D., Randall-Demllo, Sarron, Sohal, Sukhwinder Singh, Stewart, Niall, Peterson, Gregory M., Gueven, Nuri, Patel, Rahul P.. Orally Administered Enoxaparin Ameliorates Acute Colitis by Reducing Macrophage-Associated Inflammatory Responses. PloS one, vol.10, no.7, e0134259-.

  31. Mei, Q, Xu, J-M, Xiang, L, Hu, Y-M, Hu, X-P, Xu, Z-W. Change of nitric oxide in experimental colitis and its inhibition by melatonin in vivo and in vitro. Postgraduate medical journal, vol.81, no.960, 667-672.

  32. Furuhashi, Takeshi, Sugitate, Kuniyo, Nakai, Takashi, Jikumaru, Yusuke, Ishihara, Genki. Rapid profiling method for mammalian feces short chain fatty acids by GC-MS. Analytical biochemistry, vol.543, 51-54.

  33. Vemuri, Ravichandra, Shinde, Tanvi, Gundamaraju, Rohit, Gondalia, Shakuntla V., Karpe, Avinash V., Beale, David J., Martoni, Christopher J., Eri, Rajaraman. Lactobacillus acidophilus DDS-1 Modulates the Gut Microbiota and Improves Metabolic Profiles in Aging Mice. Nutrients, vol.10, no.9, 1255-.

  34. Sansone The metabolomics standards initiative Nat. Biotechnol. 2007 10.1038/nbt0807-846b 25 846 

  35. Smart, Kathleen F, Aggio, Raphael B M, Van Houtte, Jeremy R, Villas-B척as, Silas G. Analytical platform for metabolome analysis of microbial cells using methyl chloroformate derivatization followed by gas chromatography??mass spectrometry. Nature protocols, vol.5, no.10, 1709-1729.

  36. Srutkova, Dagmar, Schwarzer, Martin, Hudcovic, Tomas, Zakostelska, Zuzana, Drab, Vladimir, Spanova, Alena, Rittich, Bohuslav, Kozakova, Hana, Schabussova, Irma. Bifidobacterium longum CCM 7952 Promotes Epithelial Barrier Function and Prevents Acute DSS-Induced Colitis in Strictly Strain-Specific Manner. PloS one, vol.10, no.7, e0134050-.

  37. Han, Feifei, Fan, Hanxue, Yao, Ming, Yang, Shasha, Han, Jianzhong. Oral administration of yeast β-glucan ameliorates inflammation and intestinal barrier in dextran sodium sulfate-induced acute colitis. Journal of functional foods : the official journal of the International Society for Nutraceuticals & Functional Foods, vol.35, 115-126.

  38. Mijan, Mohammad Al, Lim, Beong Ou. Diets, functional foods, and nutraceuticals as alternative therapies for inflammatory bowel disease: Present status and future trends. World journal of gastroenterology : WJG, vol.24, no.25, 2673-2685.

  39. Ferguson, Lynnette R., Shelling, Andrew N., Browning, Brian L., Huebner, Claudia, Petermann, Ivonne. Genes, diet and inflammatory bowel disease. Mutation research. Fundamental and molecular mechanisms of mutagenesis, vol.622, no.1, 70-83.

  40. Kanauchi, Osamu; Serizawa, Isao etc. "Germinated barley foodstuff, a prebiotic product, ameliorates inflammation of colitis through modulation of the enteric environment." Journal of gastroenterology, v.38 no.2 (2003), pp. 134-141, doi:10.1007/s005350300022.

  41. Mitsuyama,, Saiki,, Kanauchi,, Iwanaga,, Tomiyasu,, Nishiyama,, Tateishi,, Shirachi,, Ide,, Suzuki,, Noguchi,, Ikeda,, Toyonaga,, Sata,. Treatment of ulcerative colitis with germinated barley foodstuff feeding: a pilot study. Alimentary pharmacology & therapeutics, vol.12, no.12, 1225-1230.

  42. Majeed, Muhammed, Nagabhushanam, Kalyanam, Natarajan, Sankaran, Sivakumar, Arumugam, Ali, Furqan, Pande, Anurag, Majeed, Shaheen, Karri, Suresh Kumar. Bacillus coagulans MTCC 5856 supplementation in the management of diarrhea predominant Irritable Bowel Syndrome: a double blind randomized placebo controlled pilot clinical study. Nutrition journal, vol.15, no.1, 21-.

  43. Turner, Jerrold R.. Intestinal mucosal barrier function in health and disease. Nature reviews. Immunology, vol.9, no.11, 799-809.

  44. Gong, Yi, Li, Hui, Li, Yan. Effects ofBacillus subtilison Epithelial Tight Junctions of Mice with Inflammatory Bowel Disease. Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research, vol.36, no.2, 75-85.

  45. Zhang, Wei, Zhu, Yao-Hong, Zhou, Dong, Wu, Qiong, Song, Dan, Dicksved, Johan, Wang, Jiu-Feng. Oral Administration of a Select Mixture of Bacillus Probiotics Affects the Gut Microbiota and Goblet Cell Function following Escherichia coli Challenge in Newly Weaned Pigs of Genotype MUC4 That Are Supposed To Be Enterotoxigenic E. coli F4ab/ac Receptor Negative. Applied and environmental microbiology, vol.83, no.3, e02747-16-e02747-16.

  46. Xavier, R. J., Podolsky, D. K.. Unravelling the pathogenesis of inflammatory bowel disease. Nature, vol.448, no.7152, 427-434.

  47. Neurath, Markus F.. Cytokines in inflammatory bowel disease. Nature reviews. Immunology, vol.14, no.5, 329-342.

  48. Hyams, Jeffrey S., Lerer, Trudy, Griffiths, Anne, Pfefferkorn, Marian, Kugathasan, Subra, Evans, Jonathan, Otley, Anthony, Carvalho, Ryan, Mack, David, Bousvaros, Athos, Rosh, Joel, Mamula, Petar, Kay, Marsha, Crandall, Wallace, Oliva-Hemker, Maria, Keljo, David, LeLeiko, Neal, Markowitz, James. Long-term outcome of maintenance infliximab therapy in children with Crohnʼs disease :. Inflammatory bowel diseases, vol.15, no.6, 816-822.

  49. Atreya, R., Mudter, J., Finotto, S., Müllberg, J., Jostock, T., Wirtz, S., Schütz, M., Bartsch, B., Holtmann, M., Becker, C., Strand, D., Czaja, J., Schlaak, J. F., Lehr, H.A., Autschbach, F., Schürmann, G., Nishimoto, N., Yoshizaki, K., Ito, H., Kishimoto, T., Galle, P.R., Rose-John, S., Neurath, M.F.. Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: Evidence in Crohn disease and experimental colitis in vivo. Nature medicine, vol.6, no.5, 583-588.

  50. Jobin, Christian, Sartor, Balfour R.. NF-κB Signaling Proteins as Therapeutic Targets for Inflammatory Bowel Diseases :. Inflammatory bowel diseases, vol.6, no.3, 206-213.

  51. Neuman, Manuela G.. Immune dysfunction in inflammatory bowel disease. Translational research : the journal of laboratory and clinical medicine, vol.149, no.4, 173-186.

  52. Soufli Cytokines and nitric oxide in immunopathogenesis of ibd and potential therapeutic approaches New Insights into Inflammatory BOWEL disease 2016 

  53. Groschwitz, Katherine R., Hogan, Simon P.. Intestinal barrier function: Molecular regulation and disease pathogenesis. The Journal of allergy and clinical immunology : official organ of American Academy of Allergy, vol.124, no.1, 3-20.

  54. Del Giudice, Marco, Gangestad, Steven W.. Rethinking IL-6 and CRP: Why they are more than inflammatory biomarkers, and why it matters. Brain, behavior, and immunity, vol.70, 61-75.

  55. Solem, Craig A, Loftus Jr, Edward V, Tremaine, William J, Harmsen, William S, Zinsmeister, Alan R, Sandborn, William J. Correlation of C-Reactive Protein with Clinical, Endoscopic, Histologic, and Radiographic Activity in Inflammatory Bowel Disease. Inflammatory bowel diseases, vol.11, no.8, 707-712.

  56. Kerr, T. A., Ciorba, M. A., Matsumoto, H., Davis, V. R.T., Luo, J., Kennedy, S., Xie, Y., Shaker, A., Dieckgraefe, B. K., Davidson, N. O.. Dextran Sodium Sulfate Inhibition of Real-Time Polymerase Chain Reaction Amplification: A Poly-A Purification Solution :. Inflammatory bowel diseases, vol.18, no.2, 344-348.

  57. Levy, Maayan, Thaiss, Christoph A., Elinav, Eran. Metabolites: messengers between the microbiota and the immune system. Genes & development, vol.30, no.14, 1589-1597.

  58. Vernocchi, Pamela, Del Chierico, Federica, Putignani, Lorenza. Gut Microbiota Profiling: Metabolomics Based Approach to Unravel Compounds Affecting Human Health. Frontiers in microbiology, vol.7, 1144-.

  59. Martin, Francois-Pierre, Su, Ming-Ming, Xie, Guo-Xiang, Guiraud, Seu Ping, Kussmann, Martin, Godin, Jean-Philippe, Jia, Wei, Nydegger, Andreas. Urinary metabolic insights into host-gut microbial interactions in healthy and IBD children. World journal of gastroenterology : WJG, vol.23, no.20, 3643-3654.

  60. Marchesi, J. R., Holmes, E., Khan, F., Kochhar, S., Scanlan, P., Shanahan, F., Wilson, I. D., Wang, Y.. Rapid and Noninvasive Metabonomic Characterization of Inflammatory Bowel Disease. Journal of proteome research, vol.6, no.2, 546-551.

  61. Robinson, Ainsley M., Gondalia, Shakuntla V., Karpe, Avinash V., Eri, Rajaraman, Beale, David J., Morrison, Paul D., Palombo, Enzo A., Nurgali, Kulmira. Fecal Microbiota and Metabolome in a Mouse Model of Spontaneous Chronic Colitis: Relevance to Human Inflammatory Bowel Disease. Inflammatory bowel diseases, vol.22, no.12, 2767-2787.

  62. Sun, Mingming, Wu, Wei, Liu, Zhanju, Cong, Yingzi. Microbiota metabolite short chain fatty acids, GPCR, and inflammatory bowel diseases. Journal of gastroenterology, vol.52, no.1, 1-8.

  63. Breuer, Richard I., Buto, Stephen K., Christ, Miriam L., Bean, Judy, Vernia, Piero, Paoluzi, P., Di Paolo, M. C., Caprilli, Renzo. Rectal irrigation with short-chain fatty acids for distal ulcerative colitis : Preliminary report. Digestive diseases and sciences, vol.36, no.2, 185-187.

  64. Koh, Ara, De Vadder, Filipe, Kovatcheva-Datchary, Petia, Bäckhed, Fredrik. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell, vol.165, no.6, 1332-1345.

  65. Kaur, Amandeep, Tuncil, Yunus E., Sikaroodi, Masoumeh, Gillevet, Patrick, Patterson, John A., Keshavarzian, Ali, Hamaker, Bruce R.. Alterations in the amounts of microbial metabolites in different regions of the mouse large intestine using variably fermentable fibres. Bioactive Carbohydrates and Dietary Fibre, vol.13, 7-13.

  66. Pang, Wanyong, Vogensen, Finn Kvist, Nielsen, Dennis Sandris, Hansen, Axel Kornerup. Faecal and caecal microbiota profiles of mice do not cluster in the same way. Laboratory animals, vol.46, no.3, 231-236.

  67. Hirano, Atsushi, Umeno, Junji, Okamoto, Yasuharu, Shibata, Hiroki, Ogura, Yoshitoshi, Moriyama, Tomohiko, Torisu, Takehiro, Fujioka, Shin, Fuyuno, Yuta, Kawarabayasi, Yutaka, Matsumoto, Takayuki, Kitazono, Takanari, Esaki, Motohiro. Comparison of the microbial community structure between inflamed and non‐inflamed sites in patients with ulcerative colitis. Journal of gastroenterology and hepatology, vol.33, no.9, 1590-1597.

  68. Majeed, Muhammed, Nagabhushanam, Kalyanam, Arumugam, Sivakumar, Natarajan, Sankaran, Majeed, Shaheen, Pande, Anurag, Beede, Kirankumar, Ali, Furqan. Cranberry seed fibre: a promising prebiotic fibre and its fermentation by the probiotic Bacillus coagulans MTCC 5856. International journal of food science & technology, vol.53, no.7, 1640-1647.

  69. Majeed, Muhammed, Majeed, Shaheen, Nagabhushanam, Kalyanam, Arumugam, Sivakumar, Natarajan, Sankaran, Beede, Kirankumar, Ali, Furqan. Galactomannan from Trigonella foenumgraecum L. seed: Prebiotic application and its fermentation by the probiotic Bacillus coagulans strain MTCC 5856. Food science & nutrition, vol.6, no.3, 666-673.

  70. Nielsen, Ditte Søvsø Gundelund, Jensen, Bent Borg, Theil, Peter Kappel, Nielsen, Tina Skau, Knudsen, Knud Erik Bach, Purup, Stig. Effect of butyrate and fermentation products on epithelial integrity in a mucus-secreting human colon cell line. Journal of functional foods : the official journal of the International Society for Nutraceuticals & Functional Foods, vol.40, 9-17.

  71. Zheng, Leon, Kelly, Caleb J., Battista, Kayla D., Schaefer, Rachel, Lanis, Jordi M., Alexeev, Erica E., Wang, Ruth X., Onyiah, Joseph C., Kominsky, Douglas J., Colgan, Sean P.. Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor–Dependent Repression of Claudin-2. The journal of immunology : official journal of the American Association of Immunologists, vol.199, no.8, 2976-2984.

  72. Simeoli, Raffaele, Mattace Raso, Giuseppina, Pirozzi, Claudio, Lama, Adriano, Santoro, Anna, Russo, Roberto, Montero‐Melendez, Trinidad, Berni Canani, Roberto, Calignano, Antonio, Perretti, Mauro, Meli, Rosaria. An orally administered butyrate‐releasing derivative reduces neutrophil recruitment and inflammation in dextran sulphate sodium‐induced murine colitis. British journal of pharmacology : BJP, vol.174, no.11, 1484-1496.

  73. Van Immerseel, Filip, Ducatelle, Richard, De Vos, Martine, Boon, Nico, Van De Wiele, Tom, Verbeke, Kristin, Rutgeerts, Paul, Sas, Benedikt, Louis, Petra, Flint, Harry J.. Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel disease. Journal of medical microbiology, vol.59, no.2, 141-143.

  74. Tedelind, Sofia, Westberg, Fredrik, Kjerrulf, Martin, Vidal, Alexander. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease.. World journal of gastroenterology : WJG, vol.13, no.20, 2826-2832.

  75. Maslowski, Kendle M., Vieira, Angelica T., Ng, Aylwin, Kranich, Jan, Sierro, Frederic, Di Yu,, Schilter, Heidi C., Rolph, Michael S., Mackay, Fabienne, Artis, David, Xavier, Ramnik J., Teixeira, Mauro M., Mackay, Charles R.. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature, vol.461, no.7268, 1282-1286.

  76. Macia, Laurence, Tan, Jian, Vieira, Angelica T., Leach, Katie, Stanley, Dragana, Luong, Suzanne, Maruya, Mikako, Ian McKenzie, Craig, Hijikata, Atsushi, Wong, Connie, Binge, Lauren, Thorburn, Alison N., Chevalier, Nina, Ang, Caroline, Marino, Eliana, Robert, Remy, Offermanns, Stefan, Teixeira, Mauro M., Moore, Robert J., Flavell, Richard A., Fagarasan, Sidonia, Mackay, Charles R.. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nature communications, vol.6, 6734-.

  77. Yuille, Samantha, Reichardt, Nicole, Panda, Suchita, Dunbar, Hayley, Mulder, Imke E.. Human gut bacteria as potent class I histone deacetylase inhibitors in vitro through production of butyric acid and valeric acid. PloS one, vol.13, no.7, e0201073-.

  78. Thorburn, Alison N., Macia, L., Mackay, Charles R.. Diet, Metabolites, and ''Western-Lifestyle'' Inflammatory Diseases. Immunity, vol.40, no.6, 833-842.

  79. Nguyen, Thi Loan Anh, Vieira-Silva, Sara, Liston, Adrian, Raes, Jeroen. How informative is the mouse for human gut microbiota research?. Disease models & mechanisms : DMM, vol.8, no.1, 1-16.

섹션별 컨텐츠 바로가기

AI-Helper ※ AI-Helper는 오픈소스 모델을 사용합니다.

AI-Helper 아이콘
AI-Helper
안녕하세요, AI-Helper입니다. 좌측 "선택된 텍스트"에서 텍스트를 선택하여 요약, 번역, 용어설명을 실행하세요.
※ AI-Helper는 부적절한 답변을 할 수 있습니다.

선택된 텍스트

맨위로