請用此 Handle URI 來引用此文件:
http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/65941完整後設資料紀錄
| DC 欄位 | 值 | 語言 |
|---|---|---|
| dc.contributor.advisor | 詹東榮(Tong-Rong Jan) | |
| dc.contributor.author | Chung-Hsiung Huang | en |
| dc.contributor.author | 黃崇雄 | zh_TW |
| dc.date.accessioned | 2021-06-17T00:15:58Z | - |
| dc.date.available | 2017-07-27 | |
| dc.date.copyright | 2012-07-27 | |
| dc.date.issued | 2012 | |
| dc.date.submitted | 2012-07-03 | |
| dc.identifier.citation | 1. Chen FC, Active ingredients and identification in common Chinese herbs. China: People Health Publ. Co., 1997.
2. Wanasundera JP, Ravindran G, Nutritional assessment of yam (Dioscorea alata) tubers. Plant Foods Hum Nutr 1994;46: 33-9. 3. Choi EM, Koo SJ, Hwang JK, Immune cell stimulating activity of mucopolysaccharide isolated from yam (Dioscorea batatas). J Ethnopharmacol 2004;91: 1-6. 4. Li J, Yang D, Yu K, He J, Zhang Y, Determination of diosgenin content in medicinal plants with enzyme-linked immunosorbent assay. Planta Med 2010;76: 1915-20. 5. DerMarderosian A, Beutler JA, The review of natural products. 3rd Edn. Missouri: Facts and Comparisons, 2000. 6. Huang K, The pharmacology of Chinese herbs. 2nd Edn. New York: CRC Press, 1999. 7. Duke JA, Ayensu ES, Medicinal plants of China. Michigan: Reference Publ. Inc, 1985. 8. Dewick P, Medicinal natural products: a biosynthetic approach. England: John Wiley & Sons Ltd. West Susses, 1997. 9. Thomas SL, Chinese and related North American herbs: phytopharmacology and therapeutic values. New York: CRC Press, 2002. 10. Attele AS, Wu JA, Yuan CS, Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol 1999;58: 1685-93. 11. Roman ID, Thewles A, Coleman R, Fractionation of livers following diosgenin treatment to elevate biliary cholesterol. Biochim Biophys Acta 1995;1255: 77-81. 12. Corbiere C, Liagre B, Bianchi A, Bordji K, Dauca M, Netter P, Beneytout JL, Different contribution of apoptosis to the antiproliferative effects of diosgenin and other plant steroids, hecogenin and tigogenin, on human 1547 osteosarcoma cells. Int J Oncol 2003;22: 899-905. 13. Hu K, Dong A, Yao X, Kobayashi H, Iwasaki S, Antineoplastic agents; I. Three spirostanol glycosides from rhizomes of Dioscorea collettii var. hypoglauca. Planta Med 1996;62: 573-5. 14. Nappez C, Liagre B, Beneytout JL, Changes in lipoxygenase activities in human erythroleukemia (HEL) cells during diosgenin-induced differentiation. Cancer Lett 1995;96: 133-40. 15. Aderiye BI, Ogundana SK, Adesanya SA, Roberts MF, Antifungal properties of yam (Dioscorea alata) peel extract. Folia Microbiol (Praha) 1996;41: 407-12. 16. Araghiniknam M, Chung S, Nelson-White T, Eskelson C, Watson RR, Antioxidant activity of dioscorea and dehydroepiandrosterone (DHEA) in older humans. Life Sci 1996;59: PL147-57. 17. Hou WC, Lee MH, Chen HJ, Liang WL, Han CH, Liu YW, Lin YH, Antioxidant activities of dioscorin, the storage protein of yam (Dioscorea batatas Decne) tuber. J Agric Food Chem 2001;49: 4956-60. 18. Lee SC, Tsai CC, Chen JC, Lin CC, Hu ML, Lu S, The evaluation of reno- and hepatoprotective effects of huai-shan-yao (Rhizome Dioscoreae). Am J Chin Med 2002;30: 609-16. 19. Choi EM, Hwang JK, Enhancement of oxidative response and cytokine production by yam mucopolysaccharide in murine peritoneal macrophage. Fitoterapia 2002;73: 629-37. 20. Daireaux M, Redini F, Loyau G, Pujol JP, Effects of associated cytokines (IL-1, TNF-alpha, IFN-gamma and TGF-beta) on collagen and glycosaminoglycan production by cultured human synovial cells. Int J Tissue React 1990;12: 21-31. 21. Lin JY, Lu S, Liou YL, Liou HL, Increased IgA and IgG serum levels using a novel yam-boxthorn noodle in a BALB/c mouse model. Food Chem Toxicol 2006;44: 170-8. 22. Fu SL, Hsu YH, Lee PY, Hou WC, Hung LC, Lin CH, Chen CM, Huang YJ, Dioscorin isolated from Dioscorea alata activates TLR4-signaling pathways and induces cytokine expression in macrophages. Biochem Biophys Res Commun 2006;339: 137-44. 23. Lin PL, Lin KW, Weng CF, Lin KC, Yam storage protein dioscorins from Dioscorea alata and Dioscorea japonica exhibit distinct immunomodulatory activities in mice. J Agric Food Chem 2009;57: 4606-13. 24. Liu YW, Liu JC, Huang CY, Wang CK, Shang HF, Hou WC, Effects of oral administration of yam tuber storage protein, dioscorin, to BALB/c mice for 21-days on immune responses. J Agric Food Chem 2009;57: 9274-9. 25. Martin FW, The species of Dioscorea containing sapogenin. Economic Botany 1969;23: 373-79. 26. Cayen MN, Ferdinandi ES, Greselin E, Dvornik D, Studies on the disposition of diosgenin in rats, dogs, monkeys and man. Atherosclerosis 1979;33: 71-87. 27. Juarez-Oropeza MA, Diaz-Zagoya JC, Rabinowitz JL, In vivo and in vitro studies of hypocholesterolemic effects of diosgenin in rats. Int J Biochem 1987;19: 679-83. 28. McAnuff MA, Harding WW, Omoruyi FO, Jacobs H, Morrison EY, Asemota HN, Hypoglycemic effects of steroidal sapogenins isolated from Jamaican bitter yam, Dioscorea polygonoides. Food Chem Toxicol 2005;43: 1667-72. 29. Al-Matubsi HY, Nasrat NA, Oriquat GA, Abu-Samak M, Al-Mzain KA, Salim M, The hypocholesterolemic and antioxidative effect of dietary diosgenin and chromium chloride supplementation on high-cholesterol fed Japanese quails. Pak J Biol Sci 2011;14: 425-32. 30. Higdon K, Scott A, Tucci M, Benghuzzi H, Tsao A, Puckett A, Cason Z, Hughes J, The use of estrogen, DHEA, and diosgenin in a sustained delivery setting as a novel treatment approach for osteoporosis in the ovariectomized adult rat model. Biomed Sci Instrum 2001;37: 281-6. 31. Jiang HP, Wu YK, Zheng W, Zeng CL, Fu WW, Fan JZ, [Structure-activity relationship of diosgenin derivatives as Bcl-2 antagonists]. Yao Xue Xue Bao 2011;46: 539-47. 32. Wang Y, Zhang YJ, Gao WY, Yan LL, [Anti-tumor constituents from Paris polyphylla var. yunnanensis]. Zhongguo Zhong Yao Za Zhi 2007;32: 1425-8. 33. Yamada T, Hoshino M, Hayakawa T, Ohhara H, Yamada H, Nakazawa T, Inagaki T, Iida M, Ogasawara T, Uchida A, Hasegawa C, Murasaki G, Miyaji M, Hirata A, Takeuchi T, Dietary diosgenin attenuates subacute intestinal inflammation associated with indomethacin in rats. Am J Physiol 1997;273: G355-64. 34. Yamaguchi A, Tazuma S, Ochi H, Chayama K, Choleretic action of diosgenin is based upon the increases in canalicular membrane fluidity and transporter activity mediating bile acid independent bile flow. Hepatol Res 2003;25: 287-95. 35. Wang YJ, Pan KL, Hsieh TC, Chang TY, Lin WH, Hsu JT, Diosgenin, a plant-derived sapogenin, exhibits antiviral activity in vitro against hepatitis C virus. J Nat Prod 2011;74: 580-4. 36. Choi KW, Park HJ, Jung DH, Kim TW, Park YM, Kim BO, Sohn EH, Moon EY, Um SH, Rhee DK, Pyo S, Inhibition of TNF-alpha-induced adhesion molecule expression by diosgenin in mouse vascular smooth muscle cells via downregulation of the MAPK, Akt and NF-kappaB signaling pathways. Vascul Pharmacol 2010;53: 273-80. 37. Wu RT, Chiang HC, Fu WC, Chien KY, Chung YM, Horng LY, Formosanin-C, an immunomodulator with antitumor activity. Int J Immunopharmacol 1990;12: 777-86. 38. Chiang HC, Wang JJ, Wu RT, Immunomodulating effects of the hydrolysis products of formosanin C and beta-ecdysone from Paris formosana Hayata. Anticancer Res 1992;12: 1475-8. 39. Jan TR, Wey SP, Kuan CC, Liao MH, Wu HY, Diosgenin, a steroidal sapogenin, enhances antigen-specific IgG2a and interferon-gamma expression in ovalbumin-sensitized BALB/c mice. Planta Med 2007;73: 421-6. 40. Sampson HA, 9. Food allergy. J Allergy Clin Immunol 2003;111: S540-7. 41. Sampson HA, Update on food allergy. J Allergy Clin Immunol 2004;113: 805-19; quiz 20. 42. Venter C, Pereira B, Grundy J, Clayton CB, Arshad SH, Dean T, Prevalence of sensitization reported and objectively assessed food hypersensitivity amongst six-year-old children: a population-based study. Pediatr Allergy Immunol 2006;17: 356-63. 43. Pereira B, Venter C, Grundy J, Clayton CB, Arshad SH, Dean T, Prevalence of sensitization to food allergens, reported adverse reaction to foods, food avoidance, and food hypersensitivity among teenagers. J Allergy Clin Immunol 2005;116: 884-92. 44. Osterballe M, Hansen TK, Mortz CG, Host A, Bindslev-Jensen C, The prevalence of food hypersensitivity in an unselected population of children and adults. Pediatr Allergy Immunol 2005;16: 567-73. 45. Noma T, Yoshizawa I, Aoki K, Yamaguchi K, Baba M, Cytokine production in children outgrowing hen egg allergy. Clin Exp Allergy 1996;26: 1298-307. 46. Tiemessen MM, Van Ieperen-Van Dijk AG, Bruijnzeel-Koomen CA, Garssen J, Knol EF, Van Hoffen E, Cow's milk-specific T-cell reactivity of children with and without persistent cow's milk allergy: key role for IL-10. J Allergy Clin Immunol 2004;113: 932-9. 47. Zeiss CR, 9. Food allergy. Intern Med 1986;105: 179-84. 48. Takayama N, Igarashi O, Kweon MN, Kiyono H, Regulatory role of Peyer's patches for the inhibition of OVA-induced allergic diarrhea. Clin Immunol 2007;123: 199-208. 49. Dannaeus A, Johansson SG, Foucard T, Ohman S, Clinical and immunological aspects of food allergy in childhood. I. Estimation of IgG, IgA and IgE antibodies to food antigens in children with food allergy and atopic dermatitis. Acta Paediatr Scand 1977;66: 31-7. 50. Abbas AK, Lichtman AH, Pillai Sm, Cellular and molecular immunology: B cell activation and antibody production. 6th Edn. USA: Elsevier, 2007. 51. Metcalfe DD, Kaliner M, Donlon MA, The mast cell. Crit Rev Immunol 1981;3: 23-74. 52. Lighvani AA, Frucht DM, Jankovic D, Yamane H, Aliberti J, Hissong BD, Nguyen BV, Gadina M, Sher A, Paul WE, T-bet is rapidly induced by interferon-gamma in lymphoid and myeloid cells. Proceedings of the National Academy of Sciences 2001;98: 15137. 53. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH, A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 2000;100: 655-69. 54. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL, Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol 1986;136: 2348-57. 55. Kelso A, Th1 and Th2 subsets: paradigms lost? Immunol Today 1995;16: 374-9. 56. Kiely PD, The Th1-Th2 model--what relevance to inflammatory arthritis? Ann Rheum Dis 1998;57: 328-30. 57. Robinson DS, Th-2 cytokines in allergic disease. British medical bulletin 2000;56: 956-68. 58. Zhang DH, Yang L, Cohn L, Parkyn L, Homer R, Ray P, Ray A, Inhibition of allergic inflammation in a murine model of asthma by expression of a dominant-negative mutant of GATA-3. Immunity 1999;11: 473-82. 59. Finotto S, De Sanctis GT, Lehr HA, Herz U, Buerke M, Schipp M, Bartsch B, Atreya R, Schmitt E, Galle PR, Renz H, Neurath MF, Treatment of allergic airway inflammation and hyperresponsiveness by antisense-induced local blockade of GATA-3 expression. J Exp Med 2001;193: 1247-60. 60. Ho IC, Lo D, Glimcher LH, c-maf promotes T helper cell type 2 (Th2) and attenuates Th1 differentiation by both interleukin 4-dependent and -independent mechanisms. The Journal of experimental medicine 1998;188: 1859-66. 61. Doncarli A, Stasiuk LM, Fournier C, Abehsira-Amar O, Conversion in vivo from an early dominant Th0/Th1 response to a Th2 phenotype during the development of collagen-induced arthritis. Eur J Immunol 1997;27: 1451-8. 62. Skolnick HS, Conover-Walker MK, Koerner CB, Sampson HA, Burks W, Wood RA, The natural history of peanut allergy. J Allergy Clin Immunol 2001;107: 367-74. 63. DeKruyff RH, Rizzo LV, Umetsu DT, Induction of immunoglobulin synthesis by CD4+ T cell clones. Semin Immunol 1993;5: 421-30. 64. Ansel KM, Lee DU, Rao A, An epigenetic view of helper T cell differentiation. Nat Immunol 2003;4: 616-23. 65. Varney VA, Tabbah K, Mavroleon G, Frew AJ, Usefulness of specific immunotherapy in patients with severe perennial allergic rhinitis induced by house dust mite: a double-blind, randomized, placebo-controlled trial. Clin Exp Allergy 2003;33: 1076-82. 66. Nasser SM, Ying S, Meng Q, Kay AB, Ewan PW, Interleukin-10 levels increase in cutaneous biopsies of patients undergoing wasp venom immunotherapy. Eur J Immunol 2001;31: 3704-13. 67. Barnes PJ, Cytokine-directed therapies for asthma. J Allergy Clin Immunol 2001;108: S72-6. 68. Targan SR, Hanauer SB, van Deventer SJ, Mayer L, Present DH, Braakman T, DeWoody KL, Schaible TF, Rutgeerts PJ, A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn's disease. Crohn's Disease cA2 Study Group. N Engl J Med 1997;337: 1029-35. 69. Busse WW, Lemanske RF, Jr., Asthma. N Engl J Med 2001;344: 350-62. 70. Borish LC, Nelson HS, Corren J, Bensch G, Busse WW, Whitmore JB, Agosti JM, Efficacy of soluble IL-4 receptor for the treatment of adults with asthma. J Allergy Clin Immunol 2001;107: 963-70. 71. Nieuwenhuizen NE, Lopata AL, Fighting food allergy: current approaches. Ann N Y Acad Sci 2005;1056: 30-45. 72. Nowak-Wegrzyn A, Future approaches to food allergy. Pediatrics 2003;111: 1672-80. 73. Gregerson DS, Obritsch WF, Donoso LA, Oral tolerance in experimental autoimmune uveoretinitis. Distinct mechanisms of resistance are induced by low dose vs. high dose feeding protocols. The Journal of Immunology 1993;151: 5751. 74. Saurer L, Mueller C, T cell-mediated immunoregulation in the gastrointestinal tract. Allergy 2009;64: 505-19. 75. Zheng Y, Rudensky AY, Foxp3 in control of the regulatory T cell lineage. Nature immunology 2007;8: 457-62. 76. Xystrakis E, Urry Z, Hawrylowicz CM, Regulatory T cell therapy as individualized medicine for asthma and allergy. Current opinion in allergy and clinical immunology 2007;7: 535. 77. Izcue A, Coombes JL, Powrie F, Regulatory T cells suppress systemic and mucosal immune activation to control intestinal inflammation. Immunol Rev 2006;212: 256-71. 78. Chehade M, Mayer L, Oral tolerance and its relation to food hypersensitivities. J Allergy Clin Immunol 2005;115: 3-12; quiz 13. 79. Weiner HL, Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells. Immunol Rev 2001;182: 207-14. 80. Weiner HL, Oral tolerance: immune mechanisms and the generation of Th3-type TGF-beta-secreting regulatory cells. Microbes Infect 2001;3: 947-54. 81. Groux H, O'Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG, A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 1997;389: 737-42. 82. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M, Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 1995;155: 1151-64. 83. Shevach EM, McHugh RS, Thornton AM, Piccirillo C, Natarajan K, Margulies DH, Control of autoimmunity by regulatory T cells. Adv Exp Med Biol 2001;490: 21-32. 84. Wills-Karp M, Santeliz J, Karp CL, The germless theory of allergic disease: revisiting the hygiene hypothesis. Nat Rev Immunol 2001;1: 69-75. 85. Renz H, Blumer N, Virna S, Sel S, Garn H, The immunological basis of the hygiene hypothesis. Chem Immunol Allergy 2006;91: 30-48. 86. Tanaka K, Ishikawa H, Role of intestinal bacterial flora in oral tolerance induction. Histol Histopathol 2004;19: 907-14. 87. Bjorksten B, Sepp E, Julge K, Voor T, Mikelsaar M, Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol 2001;108: 516-20. 88. Li XM, Serebrisky D, Lee SY, Huang CK, Bardina L, Schofield BH, Stanley JS, Burks AW, Bannon GA, Sampson HA, A murine model of peanut anaphylaxis: T- and B-cell responses to a major peanut allergen mimic human responses. J Allergy Clin Immunol 2000;106: 150-8. 89. Shida K, Makino K, Morishita A, Takamizawa K, Hachimura S, Ametani K, Sato T, Kumagai Y, Habu S, Kaminogawa S, Lactobacillus casei inhibits antigen-induced IgE secretion through regulation of cytokine production in murine splenocyte cultures. International archives of allergy and immunology 1998;115: 278-87. 90. Sanfilippo L, Li CK, Seth R, Balwin TJ, Menozzi MG, Mahida YR, Bacteroides fragilis enterotoxin induces the expression of IL-8 and transforming growth factor-beta (TGF-beta) by human colonic epithelial cells. Clin Exp Immunol 2000;119: 456-63. 91. Gibson GR, Roberfroid MB, Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr 1995;125: 1401-12. 92. Gomez-Gil B, Roque A, Turnbull JF, Inglis V, A review on the use of microorganisms as probiotics. Rev Latinoam Microbiol 1998;40: 166-72. 93. Shida K, Takahashi R, Iwadate E, Takamizawa K, Yasui H, Sato T, Habu S, Hachimura S, Kaminogawa S, Lactobacillus casei strain Shirota suppresses serum immunoglobulin E and immunoglobulin G1 responses and systemic anaphylaxis in a food allergy model. Clin Exp Allergy 2002;32: 563-70. 94. Karimi K, Inman MD, Bienenstock J, Forsythe P, Lactobacillus reuteri-induced regulatory T cells protect against an allergic airway response in mice. Am J Respir Crit Care Med 2009;179: 186-93. 95. Niers LE, Hoekstra MO, Timmerman HM, van Uden NO, de Graaf PM, Smits HH, Kimpen JL, Rijkers GT, Selection of probiotic bacteria for prevention of allergic diseases: immunomodulation of neonatal dendritic cells. Clin Exp Immunol 2007;149: 344-52. 96. Cebra JJ, Influences of microbiota on intestinal immune system development. Am J Clin Nutr 1999;69: 1046S-51S. 97. Kaila M, Isolauri E, Soppi E, Virtanen E, Laine S, Arvilommi H, Enhancement of the circulating antibody secreting cell response in human diarrhea by a human Lactobacillus strain. Pediatr Res 1992;32: 141-4. 98. Malin M, Verronen P, Korhonen H, Syvaoja EL, Salminen S, Mykkanen H, Arvilommi H, Eerola E, Isolauri E, Dietary therapy with Lactobacillus GG, bovine colostrum or bovine immune colostrum in patients with juvenile chronic arthritis: evaluation of effect on gut defence mechanisms. Inflammopharmacology 1997;5: 219-36. 99. Hoarau C, Lagaraine C, Martin L, Velge-Roussel F, Lebranchu Y, Supernatant of Bifidobacterium breve induces dendritic cell maturation, activation, and survival through a Toll-like receptor 2 pathway. J Allergy Clin Immunol 2006;117: 696-702. 100. Kruisselbrink A, Heijne Den Bak-Glashouwer MJ, Havenith CE, Thole JE, Janssen R, Recombinant Lactobacillus plantarum inhibits house dust mite-specific T-cell responses. Clin Exp Immunol 2001;126: 2-8. 101. Sistek D, Kelly R, Wickens K, Stanley T, Fitzharris P, Crane J, Is the effect of probiotics on atopic dermatitis confined to food sensitized children? Clin Exp Allergy 2006;36: 629-33. 102. Salminen SJ, Gueimonde M, Isolauri E, Probiotics that modify disease risk. J Nutr 2005;135: 1294-8. 103. Viljanen M, Savilahti E, Haahtela T, Juntunen-Backman K, Korpela R, Poussa T, Tuure T, Kuitunen M, Probiotics in the treatment of atopic eczema/dermatitis syndrome in infants: a double-blind placebo-controlled trial. Allergy 2005;60: 494-500. 104. Isolauri E, Rautava S, Kalliomaki M, Kirjavainen P, Salminen S, Role of probiotics in food hypersensitivity. Curr Opin Allergy Clin Immunol 2002;2: 263-71. 105. Di Felice G, Barletta B, Butteroni C, Corinti S, Tinghino R, Colombo P, Boirivant M, Use of probiotic bacteria for prevention and therapy of allergic diseases: studies in mouse model of allergic sensitization. J Clin Gastroenterol 2008;42 Suppl 3 Pt 1: S130-2. 106. Blumer N, Sel S, Virna S, Patrascan CC, Zimmermann S, Herz U, Renz H, Garn H, Perinatal maternal application of Lactobacillus rhamnosus GG suppresses allergic airway inflammation in mouse offspring. Clin Exp Allergy 2007;37: 348-57. 107. Ozdemir O, Various effects of different probiotic strains in allergic disorders: an update from laboratory and clinical data. Clinical & Experimental Immunology 2010;160: 295-304. 108. Su P, Henriksson A, Mitchell H, Prebiotics enhance survival and prolong the retention period of specific probiotic inocula in an in vivo murine model. J Appl Microbiol 2007;103: 2392-400. 109. Fujitani S, Ueno K, Kamiya T, Tsukahara T, Ishihara K, Kitabayashi T, Itabashi K, Increased number of CCR4-positive cells in the duodenum of ovalbumin-induced food allergy model Nc/jic mice and antiallergic activity of fructooligosaccharides. Allergol Int 2007;56: 131-8. 110. Pirapatdit S, Kishino E, Fujita K, Hashimoto H, Mori S, Saito S, Sato T, Watanabe J, Sonoyama K, Dietary alpha-linked galacto-oligosaccharide suppresses ovalbumin-induced allergic peritonitis in BALB/c mice. Biosci Biotechnol Biochem 2008;72: 1901-7. 111. Moro G, Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G, A mixture of prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the first six months of age. Arch Dis Child 2006;91: 814-9. 112. Swennen K, Courtin CM, Delcour JA, Non-digestible oligosaccharides with prebiotic properties. Crit Rev Food Sci Nutr 2006;46: 459-71. 113. Brandt EB, Strait RT, Hershko D, Wang Q, Muntel EE, Scribner TA, Zimmermann N, Finkelman FD, Rothenberg ME, Mast cells are required for experimental oral allergen-induced diarrhea. J Clin Invest 2003;112: 1666-77. 114. Drachenberg CB, Papadimitriou JC, Balaton AJ, Vaury P, The total test approach to standardization of immunohistochemistry. Arch Pathol Lab Med 2001;125: 471. 115. O'Leary TJ, Standardization in immunohistochemistry. Appl Immunohistochem Mol Morphol 2001;9: 3-8. 116. Wang CJ, Zhou ZG, Holmqvist A, Zhang H, Li Y, Adell G, Sun XF, Survivin expression quantified by Image Pro-Plus compared with visual assessment. Appl Immunohistochem Mol Morphol 2009;17: 530-5. 117. Harmsen HJ, Gibson GR, Elfferich P, Raangs GC, Wildeboer-Veloo AC, Argaiz A, Roberfroid MB, Welling GW, Comparison of viable cell counts and fluorescence in situ hybridization using specific rRNA-based probes for the quantification of human fecal bacteria. FEMS Microbiol Lett 2000;183: 125-9. 118. Wang X, Gibson GR, Effects of the in vitro fermentation of oligofructose and inulin by bacteria growing in the human large intestine. J Appl Bacteriol 1993;75: 373-80. 119. De Man JC, Rogosa M, Sharpe ME, A medium for the cultivation of lactobacilli. Journal of Applied Microbiology 1960;23: 130-35. 120. Edwards U, Rogall T, Blocker H, Emde M, Bottger EC, Isolation and direct complete nucleotide determination of entire genes. Characterization of a gene coding for 16S ribosomal RNA. Nucleic Acids Res 1989;17: 7843-53. 121. Borish L, Joseph BZ, Inflammation and the allergic response. Med Clin North Am 1992;76: 765-87. 122. Li XM, Schofield BH, Huang CK, Kleiner GI, Sampson HA, A murine model of IgE-mediated cow's milk hypersensitivity. J Allergy Clin Immunol 1999;103: 206-14. 123. Ganeshan K, Neilsen CV, Hadsaitong A, Schleimer RP, Luo X, Bryce PJ, Impairing oral tolerance promotes allergy and anaphylaxis: a new murine food allergy model. J Allergy Clin Immunol 2009;123: 231-38 e4. 124. Cardoso CR, Teixeira G, Provinciatto PR, Godoi DF, Ferreira BR, Milanezi CM, Ferraz DB, Rossi MA, Cunha FQ, Silva JS, Modulation of mucosal immunity in a murine model of food-induced intestinal inflammation. Clin Exp Allergy 2008;38: 338-49. 125. Frossard CP, Tropia L, Hauser C, Eigenmann PA, Lymphocytes in Peyer patches regulate clinical tolerance in a murine model of food allergy. J Allergy Clin Immunol 2004;113: 958-64. 126. Uhlig HH, Coombes J, Mottet C, Izcue A, Thompson C, Fanger A, Tannapfel A, Fontenot JD, Ramsdell F, Powrie F, Characterization of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during cure of colitis. J Immunol 2006;177: 5852-60. 127. Izcue A, Powrie F, Special regulatory T-cell review: Regulatory T cells and the intestinal tract-patrolling the frontier. Immunology 2008;123: 6-10. 128. Barnes MJ, Powrie F, Hybrid Treg cells: steel frames and plastic exteriors. Nat Immunol 2009;10: 563-4. 129. Zhou L, Chong MM, Littman DR, Plasticity of CD4+ T cell lineage differentiation. Immunity 2009;30: 646-55. 130. Stock P, Akbari O, Berry G, Freeman GJ, Dekruyff RH, Umetsu DT, Induction of T helper type 1-like regulatory cells that express Foxp3 and protect against airway hyper-reactivity. Nat Immunol 2004;5: 1149-56. 131. Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ, The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol 2009;10: 595-602. 132. Perelmuter K, Fraga M, Zunino P, In vitro activity of potential probiotic Lactobacillus murinus isolated from the dog. J Appl Microbiol 2008;104: 1718-25. 133. Nardi RM, Santoro MM, Oliveira JS, Pimenta AM, Ferraz VP, Benchetrit LC, Nicoli JR, Purification and molecular characterization of antibacterial compounds produced by Lactobacillus murinus strain L1. J Appl Microbiol 2005;99: 649-56. 134. Walsh MC, Gardiner GE, Hart OM, Lawlor PG, Daly M, Lynch B, Richert BT, Radcliffe S, Giblin L, Hill C, Fitzgerald GF, Stanton C, Ross P, Predominance of a bacteriocin-producing Lactobacillus salivarius component of a five-strain probiotic in the porcine ileum and effects on host immune phenotype. FEMS Microbiol Ecol 2008;64: 317-27. 135. Marcinkiewicz J, Ciszek M, Bobek M, Strus M, Heczko PB, Kurnyta M, Biedron R, Chmielarczyk A, Differential inflammatory mediator response in vitro from murine macrophages to lactobacilli and pathogenic intestinal bacteria. Int J Exp Pathol 2007;88: 155-64. 136. Reuter G, Present and future of probiotics in Germany and in Central Europe. Biosci Microflora 1997;16: 43-51. 137. Weizman Z, Asli G, Alsheikh A, Effect of a probiotic infant formula on infections in child care centers: comparison of two probiotic agents. Pediatrics 2005;115: 5-9. 138. Krasse P, Carlsson B, Dahl C, Paulsson A, Nilsson A, Sinkiewicz G, Decreased gum bleeding and reduced gingivitis by the probiotic Lactobacillus reuteri. Swed Dent J 2006;30: 55-60. 139. Forsythe P, Inman MD, Bienenstock J, Oral treatment with live Lactobacillus reuteri inhibits the allergic airway response in mice. Am J Respir Crit Care Med 2007;175: 561-9. 140. Liu Y, Fatheree NY, Mangalat N, Rhoads JM, Human-derived probiotic Lactobacillus reuteri strains differentially reduce intestinal inflammation. Am J Physiol Gastrointest Liver Physiol 2010;299: G1087-96. 141. Lee YM, Jeong HJ, Na HJ, Ku JY, Kim DK, Moon G, Chae HJ, Kim HR, Kim HM, Inhibition of immunologic and nonimmunologic stimulation-mediated anaphylactic reactions by water extract of white eggplant (Solanum melongena). Pharmacol Res 2001;43: 405-9. 142. Barrangou R, Azcarate-Peril MA, Duong T, Conners SB, Kelly RM, Klaenhammer TR, Global analysis of carbohydrate utilization by Lactobacillus acidophilus using cDNA microarrays. Proc Natl Acad Sci U S A 2006;103: 3816-21. 143. Cid SB, Miguelez-Arrizado MJ, Becker B, Holzapfel WH, Vidal-Carou MC, Amino acid decarboxylation by Lactobacillus curvatus CTC273 affected by the pH and glucose availability. Food Microbiol 2008;25: 269-77. 144. Dal Bello F, Walter J, Roos S, Jonsson H, Hertel C, Inducible gene expression in Lactobacillus reuteri LTH5531 during type II sourdough fermentation. Appl Environ Microbiol 2005;71: 5873-8. 145. Ostlie HM, Helland MH, Narvhus JA, Growth and metabolism of selected strains of probiotic bacteria in milk. Int J Food Microbiol 2003;87: 17-27. 146. Plumed-Ferrer C, Koistinen KM, Tolonen TL, Lehesranta SJ, Karenlampi SO, Makimattila E, Joutsjoki V, Virtanen V, von Wright A, Comparative study of sugar fermentation and protein expression patterns of two Lactobacillus plantarum strains grown in three different media. Appl Environ Microbiol 2008;74: 5349-58. 147. Smeianov VV, Wechter P, Broadbent JR, Hughes JE, Rodriguez BT, Christensen TK, Ardo Y, Steele JL, Comparative high-density microarray analysis of gene expression during growth of Lactobacillus helveticus in milk versus rich culture medium. Appl Environ Microbiol 2007;73: 2661-72. 148. Tymczyszyn EE, del Rosario Diaz M, Gomez-Zavaglia A, Disalvo EA, Volume recovery, surface properties and membrane integrity of Lactobacillus delbrueckii subsp. bulgaricus dehydrated in the presence of trehalose or sucrose. J Appl Microbiol 2007;103: 2410-9. 149. Saulnier DM, Molenaar D, de Vos WM, Gibson GR, Kolida S, Identification of prebiotic fructooligosaccharide metabolism in Lactobacillus plantarum WCFS1 through microarrays. Appl Environ Microbiol 2007;73: 1753-65. 150. Goh YJ, Lee JH, Hutkins RW, Functional analysis of the fructooligosaccharide utilization operon in Lactobacillus paracasei 1195. Appl Environ Microbiol 2007;73: 5716-24. | |
| dc.identifier.uri | http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/65941 | - |
| dc.description.abstract | 傳統中醫藥典籍及科學文獻記載山藥具有改善免疫及消化功能之效用,本研究之目的在於利用小鼠食物過敏模式,探討山藥中主要免疫活性成份之一〝山藥皂苷元〞的抗過敏及免疫調節作用。BALB/c小鼠分為無處理對照組、每天胃內灌服對照溶劑或山藥皂苷元之組別。除無處理對照組,其他組別小鼠皆以腹腔注射免疫卵白蛋白,並反覆胃內灌服卵白蛋白以誘發腸道過敏反應。口服給予山藥皂苷元顯著減緩卵白蛋白所誘發之腸道發炎,包括過敏性下痢的發生、杯狀細胞黏液的分泌及肥大細胞的浸潤和去顆粒。山藥皂苷元對於卵白蛋白所誘發腸道腺窩增生具減緩及保護之效用。給予山藥皂苷元可促進血清中卵白蛋白專一性抗體IgG2a生成,並促進卵白蛋白所刺激之脾臟細胞分泌細胞激素IFN-γ。然而,抗體IgE及細胞激素IL-4之生成則受到抑制。以免疫組織化學染色及逆轉錄聚合酶鏈式反應評估腸道免疫反應。結果顯示,給予山藥皂苷元可抑制IL-4及GATA3,但促進IFN-γ、T-bet、Foxp3及IL-10之表現。免疫組織化學雙染結果顯示,給予山藥皂苷元可有效回復十二指腸中因食物過敏所造成Foxp3+細胞數量之下降,並促進Foxp3+細胞分泌IL-10,但不影響TGF-β生成。以上結果證實,山藥皂苷元抗過敏之效用與促使Th1/Th2免疫平衡朝Th1為主之免疫反應及增強腸道發炎處調節型T細胞免疫反應相關。
基於山藥皂苷元與某些已報告之益生菌及益生素所表現之免疫調節效用相似,本研究分別以離體及活體試驗測試山藥皂苷元促腸內益生菌生長之效用。口服給予山藥皂苷元可有效回復因食物過敏所造成糞便中乳酸菌密度之下降。Lactobacillus murinus及L. reuteri為餵予山藥皂苷元小鼠糞便中所分離之主要乳酸菌。山藥皂苷元及其他結構相似之固醇類化合物對於此兩株乳酸菌之影響則以離體試驗測試。山藥皂苷元的添加可促進L. murinus及L. reuteri生長,但不影響enterococcus。結構活性關係分析顯示,C5-C6雙鍵及完整E和F環之結構可能為固醇類皂苷元之益生素活性所必需。總結可知,固醇類皂苷元為一可能之新型益生素。 本研究確認了山藥皂苷元抗過敏之效用並探討其可能之機制。實驗結果對於山藥改善消化及免疫系統之效用提供了科學證據,亦顯示山藥皂苷元為山藥中主要具免疫活性植物生化素之一。山藥皂苷元及其結構相似皂苷元之益生素活性未來具有應用及開發之潛力。 | zh_TW |
| dc.description.abstract | The beneficial effects of the Chinese yam to improve the function of the immune and gastrointestinal systems have been documented in the literature of traditional Chinese medicine and previous studies. The current study aimed to investigate the anti-allergic and immunomodulatory effects of diosgenin, the major immunoactive sapogenin contained in the Chinese yam, using a murine model of food allergy. BALB/c mice were either left untreated or administered daily with vehicle and/or diosgenin by gavage throughout the experiment. Except for the naive group, the mice were sensitized with ovalbumin (OVA) and repeatedly challenged with intragastric OVA to induce intestinal allergic responses. Oral administration with diosgenin markedly attenuated OVA-induced intestinal inflammation, including allergic diarrhea, mucin secretion by goblet cells, mast cell infiltration and degranulation. A protective effect by diosgenin on reducing the crypt depth of the intestine was also observed in OVA-sensitized and challenged mice. The serum production of OVA-specific IgG2a and the secretion of IFN-γ by splenocytes re-stimulated with OVA were augmented, whereas the production of IgE and IL-4 was attenuated by diosgenin administration. The immune profile in the duodenum was further characterized. The results of immunohistochemical (IHC) staining and reverse transcription-polymerase chain reaction (RT-PCR) showed that diosgenin administration suppressed the expression of IL-4 and GATA3, but enhanced the expression of IFN-γ, T-bet, Foxp3 and IL-10. The results of IHC double staining revealed that diosgenin treatment restored the diminished number of Foxp3+ cells associated with food allergy and elevated IL-10 secretion by Foxp3+ cells in the duodenum, although the production of TGF-β was unaltered. These results clearly demonstrated the anti-allergic effects of diosgenin, which is closely associated with a polarized T helper (Th)1/Th2 immunobalance toward the Th1-dominant direction and an up-regulation of the regulatory T-cell immunity in the intestinal inflammatory sites.
As the profile of diosgenin-mediated immunomodulatory effects resembles that of certain prebiotics and probiotics reported in the literature, the effect of diosgenin on the growth of enteric flora was examined both in vitro and in vivo. Oral administration with diosgenin markedly restored the diminished density of fecal lactic acid bacteria (LAB) associated with allergic reactions induced by OVA. Lactobacillus murinus and L. reuteri were the major fecal LAB isolated from diosgenin-administered mice. The direct effect of diosgenin and several structure-related steroidal compounds on the growth of the two identified LAB was investigated in vitro. The presence of diosgenin significantly enhanced the growth of both L. murinus and L. reuteri, but not enterococci. Structure–activity relationship analysis showed that the prebiotic activity of steroidal sapogenins on LAB might require structural elements of the C5–C6 double bond and intact E- and F-rings. These results indicate that steroidal sapogenins may be a novel class of prebiotics. Collectively, the current study characterized the anti-allergic effect of diosgenin and elucidated its possible underlying mechanisms. The results provide scientific evidence to substantiate the beneficial effects of the Chinese yam on the gastrointestinal and immune systems, and indicate diosgenin as one of the major immunoactive phytochemicals contained in the Chinese yam. The prebiotic potential of diosgenin and related sapogenins warrants further investigation and exploitation. | en |
| dc.description.provenance | Made available in DSpace on 2021-06-17T00:15:58Z (GMT). No. of bitstreams: 1 ntu-101-F96629005-1.pdf: 7834439 bytes, checksum: 667fda110d1dd7f779cf95ffb41502f0 (MD5) Previous issue date: 2012 | en |
| dc.description.tableofcontents | 口試委員會審定書 #
中文摘要 ii ABSTRACT iv CONTENTS vii LIST OF FIGURES xii LIST OF TABLES xiv ABBREVIATIONS xv Chapter 1 Introduction 1 1.1 The Chinese yam 1 1.1.1 Background of the Chinese yam 1 1.1.2 Major constituents of yams 1 1.1.3 Therapeutic effects of the Chinese yam in traditional Chinese medicine 2 1.1.4 Biological activities of the Chinese yam 2 1.2 Diosgenin 4 1.2.1 Background of diosgenin 4 1.2.2 Biological activities of diosgenin 5 1.2.2.1 Anti-inflammatory effect 5 1.2.2.2 Anti-osteoporosis effect 6 1.2.2.3 Anti-oxidative and hypocholesterolemic effect 6 1.2.2.4 Anti-tumor effect 7 1.2.2.5 Anti-viral effect 7 1.2.2.6 Hypoglycemic effect 8 1.2.2.7 Immunomodulatory effect 8 1.3 Food allergy 9 1.3.1 Background of food allergy 9 1.3.2 Immunopathology of food allergy 10 1.3.3 Th1/Th2 immune balance 10 1.3.4 Strategies for Therapies of food allergy 12 1.4 Regulatory T cell (Treg cell) 13 1.4.1 Role of Treg cell in the gastrointestinal immunity 13 1.5 Probiotics and prebiotics 14 1.5.1 Role of enteric bacteria in allergy 14 1.5.2 Potential therapeutic effects of probiotics on allergy 15 1.5.3 Potential therapeutic effects of prebiotics on allergy 16 1.6 Hypothesis and objective of this study 18 1.7 Graphic abstract of this dissertation research 21 Chapter 2 Materials and methods 22 2.1 Chemicals and reagents 22 2.2 Murine model of food allergy 23 2.2.1 Animals 23 2.2.2 Protocol of animal experiments 23 2.3 Necropsy and intestinal tissue preparation 26 2.4 Culture of splenocytes re-stimulated with ovalbumin (OVA) 26 2.5 Enzyme-linked immunosorbent assay (ELISA) for serum antibodies and cytokines detection 27 2.6 Morphometric analysis 28 2.7 Immunohistochemical staining (IHC) 28 2.7.1 IHC single staining 28 2.7.2 IHC double staining 29 2.7.3 Quantification of IHC staining 30 2.8 Reverse Transcription-Polymerase Chain Reaction (RT-PCR) 30 2.9 Fecal bacteria analysis 33 2.9.1 Culture of fecal bacteria 33 2.9.2 Identification of isolated fecal microbes 33 2.9.3 In vitro growth assay of isolated fecal microbes 34 2.10 Statistical analysis 34 Chapter 3 Experimental results 36 3.1 Anti-allergic effects of diosgenin 36 3.1.1 Diosgenin administration attenuated the occurrence of allergic diarrhea 36 3.1.2 Diosgenin administration attenuated allergen-induced intestinal inflammation 38 3.1.3 Diosgenin administration suppressed total and OVA-specific IgE production whereas enhanced OVA-specific IgG2a production in serum 44 3.1.4 Diosgenin administration enhanced IFN-γ secretion whereas inhibited IL-4 secretion by splenocytes 46 3.2 Modulatory effects of diosgenin on the intestinal immunity 47 3.2.1 Diosgenin administration skewed Th1/Th2 immune balance toward Th1 direction in the duodenum 47 3.2.2 Diosgenin administration restored the depleted number of Foxp3+ Treg cells and enhanced IL-10 production in the duodenum 53 3.3 In vivo and in vitro prebiotic activity of diosgenin 55 3.3.1 Diosgenin administration restored the depleted density of lactic acid bacteria and suppressed the elevated density of non-specific bacteria in feces 55 3.3.2 In vitro diosgenin treatment enhanced the growth of Lactobacillus murinus and L. reuteri 58 3.3.3 Structure–activity relationship (SAR) analysis on the prebiotic activity of steroidal sapogenins 61 Chapter 4 Discussion 65 REFERENCES 74 | |
| dc.language.iso | en | |
| dc.subject | T細胞 | zh_TW |
| dc.subject | 山藥皂苷 | zh_TW |
| dc.subject | 元 | zh_TW |
| dc.subject | 食物過敏 | zh_TW |
| dc.subject | 免疫 | zh_TW |
| dc.subject | 益生素 | zh_TW |
| dc.subject | 山藥 | zh_TW |
| dc.subject | the Chinese yam | en |
| dc.subject | food allergy | en |
| dc.subject | immune | en |
| dc.subject | diosgenin | en |
| dc.subject | prebiotics | en |
| dc.subject | T cell | en |
| dc.title | 山藥皂苷元抗過敏及益生素之效用 | zh_TW |
| dc.title | Anti-allergic and prebiotic effects of diosgenin, the major steroidal sapogenin derived from the Chinese yam | en |
| dc.type | Thesis | |
| dc.date.schoolyear | 100-2 | |
| dc.description.degree | 博士 | |
| dc.contributor.oralexamcommittee | 沈立言,周崇熙,簡茂盛,梁弘人,林俊宏 | |
| dc.subject.keyword | 山藥皂苷,元,食物過敏,免疫,益生素,山藥,T細胞, | zh_TW |
| dc.subject.keyword | diosgenin,food allergy,immune,prebiotics,the Chinese yam,T cell, | en |
| dc.relation.page | 97 | |
| dc.rights.note | 有償授權 | |
| dc.date.accepted | 2012-07-04 | |
| dc.contributor.author-college | 獸醫專業學院 | zh_TW |
| dc.contributor.author-dept | 獸醫學研究所 | zh_TW |
| 顯示於系所單位: | 獸醫學系 | |
文件中的檔案:
| 檔案 | 大小 | 格式 | |
|---|---|---|---|
| ntu-101-1.pdf 未授權公開取用 | 7.65 MB | Adobe PDF |
系統中的文件,除了特別指名其著作權條款之外,均受到著作權保護,並且保留所有的權利。
