Gruger Family Fungi

Check out our site, subscribe to my channel here on youtube for more goodies! Plus follow us on facebook for more news and regular posts on updates.
instagram.com/gruger_market/
facebook.com/grugermarket/
twitter.com/gruger_market
www.grugermarket.ca
[email protected]

Reishi: 157 References 1. Hobbs C. Medicinal Mushrooms. Santa Cruz: Botanica Press; 1995. 2. Lelley J. Die Heilkraft der Pilze. Berlin: ECON-Verlag; 1997. 3. Lindequist U. Ganoderma. In: Schneider G, Hänsel R, Blaschek W, editors. HAGERs Handbuch der Pharmazeutischen Praxis. Berlin, Heidelberg, New York: Springer-Verlag; 1998. pp. 750–61. (in German) 4. Lindequist U. Lentinula. In: Schneider G, Hänsel R, Blaschek W, editors. HAGERs Handbuch der Pharmazeutischen Praxis. Berlin, Heidelberg, New York: Springer-Verlag; 1998. pp. 61–71. (in German) 5. Lindequist U. Schizophyllum. In: Schneider G, Hänsel R, Blaschek W, editors. HAGERs Handbuch der Pharmazeutischen Praxis. Berlin, Heidelberg, New York: Springer-Verlag; 1998. pp. 528–34. (in German) 6. Stamets P. Growing Gourmet and Medicinal Mushrooms. Berkely: Ten Speed Press; 2000. 7. Zjawiony J. Biologically active compounds from Aphyllophorales (Polypore) fungi. J Nat Prod.2004;67:300–10. 8. Chang ST. Mushroom research and development—equality and mutual benefit. In: Royse DJ, editor. Proceedings of the 2nd International Conference on Mushroom Biology and Mushroom Products.Pennsylvania State University; 1996. pp. 1–10. 9. Wasser SP, Nevo E, Sokolov D, Reshetnikov S. Timot-Tismenetsky M. Dietary supplements from medicinal mushrooms: diversity of types and variety of regulations. Int J Med Mushrooms. 2000;2:1–19. 10. Chang ST, Miles PG. Mushrooms biology—a new discipline. Mycologist. 1992;6:64–5. 11. Hawksworth DL. Mushrooms: the extent of the unexplored potential. Int J Med Mushrooms.2001;3:333–7. 12. Lindequist U, Teuscher E, Narbe G. Neue Wirkstoffe aus Basidiomyceten. Z Phytother. 1990;11:139–49.(in German) 13. Mothana RAA, Jansen R, Jülich W-D, Lindequist U. Ganomycin A and B, new antimicrobial farnesyl hydroquinones from the basidiomycete Ganoderma pfeifferi. J Nat Prod. 2000;63:416–8. [PubMed] 14. Smania EFA, Delle Monache F, Smania Jr A, Yunes RA. Cuneo. Antifungal activity of sterols and triterpenes isolated from Ganoderma annulare. Fitoterapia. 2003;74:375–7. [PubMed] 15. Smania A, Jr, Delle Monache F, Smania EFA, Cuneo RS. Antibacterial activity of steroidal compounds isolated from Ganoderma applanatum (Pers.) Pat. (Aphyllophoromycetideae) fruit body. Int J Med Mushrooms. 1999;1:325–30. 16. Bender S, Dumitrache CN, Backhaus J, Christie G, Cross RF, Lonergan GT, et al. A case for caution in assessing the antibiotic activity of extracts of culinary-medicinal Shiitake mushroom [Lentinus edodes(Berk.)Singer] (Agaricomycetidae) Int J Med Mushrooms. 2003;5:31–5.
17. Badalyan SM. Antiprotozoal activity and mitogenic effect of mycelium of culinary-medicinal shiitake mushroom Lentinus edodes (Berk.) Singer (Agaricomycetidae) Int J Med Mushrooms. 2004;6:131–8.
18. Al-Fatimi MAM. 2001. Isolierung und Charakterisierung antibiotisch wirksamer Verbindungen aus Ganoderma pfeifferi Bres. und aus Podaxis pistillaris (L.:Pers.) Morse. Universität Greifswald. (in German)
19. Brandt CR, Piraino F. Mushroom antivirals. Recent Res Dev Antimicrob Agents Chemother. 2000;4:11–26.
20. El-Mekkawy S, Meselhy MR, Nakamura N, Tezuka Y, Hattori M, Kakiuchi N, et al. Anti-HIV-1 and anti-HIV-1-protease substances from Ganoderma lucidum. Phytochemistry. 1998;49:1651–7.
21. Mothana RAA, Awadh NAA, Jansen R, Wegner U, Mentel R, Lindequist U. Antiviral lanostanoid triterpenes from the fungus Ganoderma pfeifferi BRES. Fitoterapia. 2003;74:177–80.
22. Mentel R, Meinsen D, Pilgrim H, Herrmann B, Lindequist U. In vitro antiviral effect of extracts of Kuehneromyces mutabilis on influenza virus. Pharmazie. 1994;49:859–60.
23. Awadh AAN, Mothana RAA, Lesnau A, Pilgrim H, Lindequist U. Antiviral activity of extracts and compounds from Inonotus hispidus. Fitotherapia. 2003;74:483–5.
24. Lindequist U, Lesnau A, Teuscher E, Pilgrim H. Untersuchungen zur antiviralen Wirksamkeit von Ergosterolperoxid. Pharmazie. 1989;44:579–80. (in German)
25. Leonhardt K, Anke T, Hillen-Maske E, Steglich W. 6-Methylpurine, 6-methyl-9-β-D-ribofuranosyl-purine, and 6-hydroxymethyl-9-β-D-ribofuranosyl-purin as antiviral metabolites of Collybia maculata(basidiomycetes) Z Naturforsch C. 1987;42:420–4.
26. Ichimura T, Watanabe O, Maruyama S. Inhibition of HIV-1 protease by water-soluble lignin-like substance from an edible mushroom, Fuscoporia obliqua. Biosci Biotechnol Biochem. 1998;62:575–7.
27. Tochikura TS, Nakashima H, Ohashi Y, Yamamoto N. Inhibition (in vitro) of replication and of the cytopathic effect of human immunodeficiency virus by an extract of the culture medium of Lentinus edodesmycelia. Med Microbiol Immunol. 1988;177:235–44.
28. Suzuki H, Okubo A, Yamazaki S, Suzuki K, Mitsuya H, Toda S. Inhibition of the infectivity and cytopathic effect of human immunodeficiency virus by water-soluble lignin in an extract of the culture medium of Lentinus edodes mycelia (LEM) Biochem Biophys Res Commun. 1989;160:367–73.
29. Yoshida O, Nakashima H, Yoshida T, Kaneko Y, Yamamoto I, Matsuzaki K, et al. Sulfation of the immunomodulating polysaccharide lentinan: a novel strategy for antivirals to human immunodeficiency virus (HIV) Biochem Pharmacol. 1988;37:2887–91.
30. Tochikura TS, Nakashima H, Hirose K, Yamamoto N. A biological response modifier, PSK, inhibits human immunodeficiency virus infection in vitro. Biochem Biophys Res Commun. 1987;148:726–33.
31. Colins RA, Ng TB. Polysaccharopeptide from Coriolus versicolor has potential for use against human immunodeficiency virus type 1 infection. Life Sci. 1997;60:PL383–7.
32. Wang HX, Ng TB. Isolation and characterization of velutin, a novel low-molecular-weight ribosome-inactivating protein from winter mushroom (Flammulina velutipes) fruiting bodies. Life Sci. 2001;68:2151–8.
33. Nanba H, Kodama N, Schar D, Turner D. Effects of maitake (Grifola frondosa) glucan in HIV-infected patients. Mycoscience. 2000;41:293–5.
34. Semerdzieva M, Veselsky J. Academia Praha; 1986. Lecive houby drive a nyni. (in Czech)
35. Molitoris HP. Mushrooms in medicine. Folia Microbiol. 1994;39:91–8.
36. Kahlos K, Kangas L, Hiltunen R. Antitumor activity of some compounds and fractions from an n-hexane extract of Inonotus obliquus in vitro. Acta Pharm Fennica. 1987;96:33–40.
37. Burczyk J, Gawron A, Slotwinska M, Smietana B, Terminska K. Antimitotic activity of aqueous extracts of Inonotus obliquus. Boll Chim Farm. 1996;135:306–9.
38. Babitskaya VG, Scherba VV, Ikonnikova NV, Bisko NA, Mitropolskaya NY. Melanin complex from medicinal mushroom Inonotus obliquus (Pers.:Fr.) Pilat (Chaga) (Aphyllophoromycetidae) Int J Med Mushrooms. 2002;4:139–45.
39. Chihara G, Maeda Y, Sasaki T, Fukuoka F. Inhibition of mouse sarcoma 180 by polysaccharides from Lentinus edodes (Berk.) Nature. 1969;222:687–8.
40. Mizuno T. The extraction and development of antitumor-active polysaccharides from medicinal mushrooms in Japan (review) Int J Med Mushrooms. 1999;1:9–30.
41. Wasser SP, Weis AL. Medicinal properties of substances occurring in higher Basidiomycetes mushrooms: current perspectives (review) Int J Med Mushrooms. 1999;1:31–62.
42. Reshetnikov SV, Wasser SP, Tan KK. Higher basidiomycetes as a source of antitumor and immunostimulating polysaccharides (review) Int J Med Mushrooms. 2001;3:361–94.
43. Zhou S, Gao Y. The immunomodulating effects of Ganoderma lucidum (Curt.:Fr.) P.Karst (LingZhi, Reishi Mushroom) (Aphylloromycetidae) Int J Med Mushrooms. 2002;4:1–11.
44. Karinaga R, Mizu M, Koumoto K, Anada T, Shinkai S, Kimura T, et al. First observation by fluorescence polarization of complexation between mRNA and the natural polysaccharide schizophyllan. Chem Biodivers. 2004;1:634–9.
45. Adachi Y, Suzuki Y, Jinushi T, Yadomae T, Ohno N. Th1-oriented immunomodulating activity of gel-forming fungal (1-3)-beta-glucans. Int J Med Mushrooms. 2002;4:95–109.
46. Nanba H, Hamaguchi A, Kuroda H. The chemical structure of an antitumor polysaccharide in fruit bodies of Grifola frondosa (maitake) Chem Pharm Bull (Tokyo) 1987;35:1162–8.
47. Kodama N, Komuta K, Nanba H. Effect of maitake (Grifola frondosa) D-fraction on the activation of NK cells in cancer patients. J Med Food. 2003;6:371–7.
48. Hazama S, Oka M, Yoshino S, Iizuka N, Wadamori K. Yamamoto, et al. Clinical effects and immunological analysis of intraabdominal and intrapleural injection of lentinan for malignant ascites and pleural effusion of gastric carcinoma. Cancer Chemother. 1995;22:1595–7.
49. Ochiai T, Isono K, Suzuki T, Koide Y, Gunji Y, Nagata M, et al. Int J Immunother. 1992;8:161–9.
50. Taguchi T, Furue H, Kimura T, Kondoh T, Hattori T, Itoh I, et al. Life-span prolongation effect of lentinan on patients with advanced or recurrent colorectal cancer. Int J Immunopharmacol. 1982;4:271.
51. Fujimoto S, Furue H, Kimura T, Kondo T, Orita K, Taguchi T, et al. Clinical outcome of postoperative adjuvant immunochemotherapy with sizofiran for patients with resectable gastric cancer—a randomised controlled study. Eur J Cancer. 1991;27:1114–8.
52. Mitomi T, Tsuchiya S, Iijima N, Aso K, Suzuki K, Nishiyama K, et al. Randomized, controlled study on adjuvant immunochemotherapy with PSK in curatively resected colorectal cancer. Dis Colon Rectum.1992;35:123–30.
53. Yang QY. A new biological response modifier – PSP. In: Chang ST, editor. Mushroom Biology and Mushroom Products. Hong Kong: The Chinese University Press; 1993. pp. 247–59.
54. Gordon M, Guralnik M, Kaneko Y, Mimura T, Goodgame J, DeMarzo C, et al. A phase II controlled study of a combination of the immune modulator, lentinan, with didanosine (DDI) in HIV patients with CD4 cells of 200–500/MM(3) J Med. 1995;26:193–207.
55. Kodama N, Komuta K, Nanba H. Can maitake MD-fraction aid cancer patients? Altern Med Rev.2002;7:236–9.
56. Konno S, Aynehchi S, Dolin DJ, Schwartz AM, Choudhury MS, Tazakin HN. Anticancer and hypoglycemic effects of polysaccharides in edible and medicinal Maitake mushroom [Grifola frondosa(Dicks.:Fr.) S.F.Gray] Int J Med Mushrooms. 2002;4:185–95.
57. Yap AT, Ng ML. Immunopotentiating properties of lentinan (1-3)- β-d-glucan extracted from culinary-medicinal Shiitake mushroom Lentinus edodes (Berk.) Singer (Agaricomycetidae) Int J Med Mushrooms.2003;5:339–58.
58. Bodinet C, Lindequist U, Teuscher E, Freudenstein J. Influence of peroral application of a herbal immunomodulator on the antibody production of Peyer’s Patches-cells. Arzneim Forsch. 2004;54:114–8.
59. Gao Y, Dai X, Chen G, Ye J, Zhou S. A randomized, placebo-controlled, multicenter study of Ganoderma lucidum (W.Curt.:Fr.) Lloyd (Aphylloromycetidae) polysaccharides (Ganopoly R) in patients with advanced lung cancer. Int J Med Mushrooms. 2003;5:369–81.
60. Gao Y, Zhou S, Chen G, Dai X, Ye J. A phase I/II study of a Ganoderma lucidum (Curt.:Fr.) P.Karst. extract (Ganopoly) in patients with advanced cancer. Int J Med Mushrooms. 2002;4:207–14.
61. Ohno N, Harada T, Masuzawa S, Miura NN, Adachi Y, Nakajima M, et al. Antitumor activity and hematopoietic response of a β-glucan extracted from an edible and medicinal mushroom Sparassis crispaWulf.:Fr. (Aphylloromycetidae) Int J Med Mushrooms. 2002;4:13–26.
62. Ohno N, Nameda S, Harada T, Miura NN, Adachi Y, Nakajima M, et al. Immunomodulating activity of a β-glucan preparation, SCG, extracted from a culinary-medicinal mushroom, Sparassis crispa Wulf.:Fr. (Aphyllophoromycetidae), and application to cancer patients. Int J Med Mushrooms. 2003;5:359–68.
63. Mizuno T. Bioactive substances in Hericium erinaceus (Bull.:Fr.) Pers. (Yamabushitake), and its medicinal utilization. Int J Med Mushrooms. 1999;1:105–19.
64. Stijve T, de A Amazonas MA, Giller V. Flavour and taste components of Agarius blazei Murrill ss. Heinem.—a new gourmet and medicinal mushroom. Dtsch Lebensm-Rundsch. 2002;98:448–53.
65. Wasser SP, Didukh MY, de A Amazonas MAL, Nevo E, Stamets P, da Eira AF. Is a widely cultivated culinary-medicinal royal sun Agaricus (the Himematsutake mushroom) indeed Agaricus blazei Murrill? Int J Med Mushrooms. 2002;4:267–90.
66. Mizuno T. Medicinal properties and clinical effects of culinary-medicinal mushroom Agaricus blazeiMurrill (Agaricomycetidae) (Review) Int J Med Mushrooms. 2002;4:299–312.
67. Yuexin L, Zhuqiu Y, Yanyan H, Hualing X. Fractionation and characterization of water-soluble polysaccharides from culinary-medicinal mushroom, Agaricus blazei Murrill (Agaricomycetidae) fruit body. Int J Med Mushrooms. 2002;4:313–9.
68. Menoli RC, Mantovani MS, Ribeiro LR, Speit G, Jordalo BQ. Antimutagenic effects of the mushroom Agaricus blazei Murrill extracts on v79 cells. Mutat Res. 2001;12:5–13.
69. Lam YW, Ng TB, Wang HX. Antiproliferative and antimitogenic activities in a peptide from puffball mushroom Calvatia caelata. Biochem Biophys Res Commun. 2001;289:744–9.
70. Lam SK, Ng TB. Hypsin, a novel thermostable ribosome inactivating protein with antifungal and antiproliferative activities from fruiting bodies of the edible mushroom Hypsizigus marmoreus. Biochem Biophys Res Commun. 2001;285:1071–5.
71. Yu LG, Fernig DJ, Smith JA, Milton JD, Rhodes JM. Reversible inhibition of proliferation of epithelial cell lines by Agaricus bisporus (edible mushroom) lectin. Cancer Res. 1993;53:4627–32.
75. Ajith TA, Janardhanan KK. Cytotoxic and antitumor activities of a polypore macrofungus, Phellinus rimosus (Berk) Pilat. J Ethnopharmacol. 2003;84:157–62.
76. McMorris TC, Kelner MJ, Wang W, Estas LA, Montoya MA, Taetle R. Structure-activity relationships of illudin analogs with improved therapeutic index. J Org Chem. 1992;57:6876–83.
77. Hartting U, Anke T, Scherer A, Steglich W. Leaianafulvene, a sesquiterpenoid fulvene derivative from culture of Mycena leaviana. Phytochemistry. 1990;29:3942–4.
78. Toth JO, Luu B, Ourisson G. Ganoderic acid T and Z: cytotoxic triterpenes from Ganoderma lucidum(Polyporaceae) Tetrahedron Lett. 1983;24:1081–4.
79. Toth JO, Luu B, Beck JP, Ourisson G. Chemistry and biochemistry of Oriental drugs. Part IX. Cytotoxic triterpenes from Ganoderma lucidum (Polyporaceae): structures of ganoderic acids U-Z. J Chem Res Synop.1983;12:299.
80. Gao JJ, Min BS, Ahn EM, Nakamura N, Lee HK, Hattori M. New triterpene aldehydes, lucialdehydes A-C, from Ganoderma lucidum and their cytotoxicity against murine and human tumor cells. Chem Pharm Bull. 2002;50:837–40.
81. Leon F, Valencia M, Augusto R, Nieto I, Quintana J, Estevez F, et al. Novel cytostatic lanostanoid triterpenes from Ganoderma australe. Helv Chim Acta. 2003;86:3088–95.
82. Nam KS, Jo YS, Kim YH, Hyun JW, Kim HW. Cytotoxic activities of acetoxyscirpenediol and ergosterol peroxide from Paecilomyces tenuipes. Life Sci. 2001;69:229–37.
83. Bok JW, Lermer L, Chilton J, Klingeman HG, Towers GHN. Antitumor sterols from the mycelia of Cordyceps sinensis. Phytochemistry. 1999;51:891–8.
84. Han HC, Lindequist U, Hyun JW, Kim YH, An HS, Lee DH, et al. Apoptosis induction by acetoxyscirpendiol from Paecilomyces tenuipes in human leukaemia cell lines. Pharmazie. 2004;59:42–9.
85. Gonzalez AG, Leon F, Rivera A, Padron JI, Gonzalez-Plata J, Zuluaga JC, et al. New lanostanoids from the fungus Ganoderma concinna. J Nat Prod. 2002;65:417–21.
86. Chairul, Tokuyama T, Hayashi Y, Nishizawa M, Tokuda H, Chairul SM, et al. Applanoxidic acids A, B, C and D, biologically active tetracyclic triterpenes from Ganoderma applanatum. Phytochemistry.1991;30:4105–9.
87. Chairul, Chairul SM, Hayashi Y. Lanostanoid triterpenes from Ganoderma applanatum. Phytochemistry.1994;35:1305–8.
88. Murgo A, Cannon DJ, Blatner G, Cheson BD. Clinical trials of MGI-114. Oncology. 1999;13:233–8.
89. Lee S, Park S, Oh JW, Yang C. Natural inhibitors for protein prenyltransferase. Planta Med.1998;64:303–8.
90. Kim SH, Song YS, Kim SK, Kim BC, Lim CJ, Park EH. Anti-inflammatory and related pharmacological activities of the n-BuOH subfraction of mushroom Phellinus linteus. J Ethnopharmacol.2004;93:141–6.
91. Cho JH, Cho SD, Hu H, Kim SH, Lee SK, Lee YS, et al. The roles of ERK 1/2 and p38 MAP kinases in the prevention mechanism of mushroom Phellinus linteus against the inhibition of gap junctional intercellular communication by hydrogen peroxide. Carcinogenesis. 2002;23:1164–9.
92. Takaku T, Kimura Y, Okuda H. Isolation of an antitumor compound from Agaricus blazei Murrill and mechanism of action. J Nutr. 2001;5:1409–13.
93. Hashimoto T, Asakawa Y. Biologically active substances of Japanese inedible mushrooms. Heterocycles.1998;47:1067–110.
95. Mlinrič A, Kac J, Fatur T, Filini M. Anti-genotoxic activity of the mushroom Lactarius vellereus extract in bacteria and in mammalian cells in vitro. Pharmazie. 2004;59:217–21.
96. Shi YL, James AE, Benzie IFF, Buswell JA. Genoprotective activity of edible and medicinal mushroom components. Int J Med Mushrooms. 2004;6:1–14.
98. Sano M, Yoshino K, Matsuzawa T, Ikekawa T. Inhibitory effects of edible higher basidiomycetes mushroom extracts on mouse type IV allergy. Int J Med Mushrooms. 2002;4:37–41.
99. Kohda H, Tokumoto W, Sakamoto K, Fujii M, Hirai Y, Yamasaki K, et al. The biologically-active constituents of Ganoderma lucidum (Fr) Karst—histamine release-inhibitory triterpenes. Chem Pharm Bull.1985;33:1367–73.
100. Tasaka K, Mio M, Izushi K, Akagi M, Makino T. Anti-allergic constituents in the culture medium of Ganoderma lucidum. (II). The inhibitory effect of cyclooctasulfur on histamine release. Agents Actions.1988;23:157–60.
101. Lindequist U, Teuscher E, Wolf B, Völsgen A, Hoffmann S, Kutschabsky L, et al. Isolierung, Charakterisierung und Strukturaufklärung eines immunsuppressiv wirksamen Inhaltsstoffes aus Tricholoma populinum LANGE. Pharmazie. 1989;44:165. (in German)
102. Kreisel H, Lindequist U, Horak M. Distribution, ecology and immunosuppressive properties of Tricholoma populinum (Basidiomycetes) Zentralbl Mikrobiol. 1990;145:393–6.
103. Ali NAA, Pilgrim H, Lüdke J, Lindequist U. Inhibition of chemiluminescence response of human mononuclear cells and suppression of mitogen-induced proliferation of spleen lymphocytes of mice by hispolon and hispidin. Pharmazie. 1996;51:667–70.]
104. Koch J, Witt S, Lindequist U. The influence of selected basidiomycetes on the binding of lipopolysaccharide to its receptor. Int J Med Mushrooms. 2002;4:229–35.
105. Min BS, Gao JJ, Hattori M, Lee HK, Kim YH. Anticomplement activity of terpenoids from the spores of Ganoderma lucidum. Planta Med. 2001;67:811–4. [PubMed]
108. Chen Q. Antilipemic effect of polysaccharides from Auricularia auricular, Tremella fuciformis, and Tremella fuciformis spores. Zhongguo Yaoke Daxue Xuebao. 1989;20:344–7.
109. Cheung PCK. The hypocholesterolemic effect of two edible mushrooms: Auricularia auricula (tree-ear) and Tremella fuciformis (white jelly-leaf) in hypercholesterolemic rats. Nutr Res. 1996;16:1721–5.
110. Komoda Y, Shimizu M, Sonoda Y, Sato Y. Ganoderic acid and its derivatives as cholesterol synthesis inhibitors. Chem Pharm Bull. 1989;37:531–3.
111. Morigiwa A, Kitabatake K, Fujimoto Y, Ihekawa N. Angiotensin converting enzyme inhibitory triterpenes from Ganoderma lucidum. Chem Pharm Bull. 1986;34:3025–8.
112. Su CY, Shiao MS, Wang CT. Predominant inhibition of ganodermic acid S on the thromboxane A2-dependent pathway in human platelets response to collagen. Biochim Biophys Acta. 1999;1437:223–34.
113. Lin ZB. Focus on anti-oxidative and free radical scavening activity of Ganoderma lucidum. J Appl Pharmacol. 2004;12:133–7.
114. Tokuda S, Tapiri A, Kano E, Sugwara Y, Suzuki S, Sato H, et al. Reducing mechanism of plasma cholesterol by Shii-ta-ke. Mushroom Sci. 1974;9:445–61.
115. Liu JK, Hu L, Dong ZJ, H Q. DPPH radical scavenging activity of ten natural p-terphenyl derivatives obtained from three edible mushrooms indigenous to China. Chem Biodivers. 2004;1:601–5.
116. Yun BS, Lee Iky, Kim JP, Yoo ID. Two p-terphenyls from mushroom Paxillus panuoides with free radical scavenging activity. J Microbiol Biotechnol. 2000;10:233–7.
117. Lee IK, Yun BS, Cho SM, Kim WG, Kim JP, Ryoo IJ, et al. Betulinans A and B, two Benzoquinone Compounds from Lenzites betulina. J Nat Prod. 1996;59:1090–2.
118. Yun BS, Cho Y, Lee IK, Cho SM, Lee TH, Yoo ID. Sterins A and B, new antioxidative compounds from Stereum hirsutum. J Antibiot. 2002;55:208–10.
119. Yuan D, Mori J, Komatsu K, Makino T, Kano Y. An anti-aldosteronic diuretic component (drain dampness) in Polyporus sclerotium. Biol Pharm Bull. 2004;27:867–70.
120. Lu W, Adachi I, Kano K, Yasuta A, Toriizuka K, Ueno M, et al. Platelet aggregation potentiators from Cho-Re. Chem Pharm Bull. 1985;33:5083–7.
121. Hikino H, Konno C, Mirin Y, Hayashi T. Isolation and hypoglycaemic activities of ganoderans A and B, glucans of Ganoderma lucidum fruit bodies. Planta Med. 1985;51:339–40.
122. Ikuzawa M, Oguchi Y, Matsunaga K, Toyoda N, Furusho T, Fujii T, et al. Pharmaceutical preparation containing a glycoprotein. German Patent DE. 3,429,551. 1985.
123. Kiho T, Morimoto H, Kobayashi T, Ysai S, Ukai S, Aizawa K, et al. Effect of a polysaccharide (TAP) from the fruiting bodies of Tremella aurantia on glucose metabolism in mouse liver. Biosci Biotechnol Biochem. 2000;64:417–9.
124. Gao Y, Lan J, Dai X, Ye J, Zhou S. A phase I/II study of ling zhi mushroom Ganoderma lucidum(W.Curt.:Fr.)Lloyd (Aphyllophoromycetidae) extract in patients with type II diabetes mellitus. Int J Med Mushrooms. 2004;6:33–9.
125. Wasser SP, Tan KK, Elisashvili VI. Hypoglycemic, interferonogenous, and immunomodulatory activity of Tremellastin from the submerged culture of Tremella mesenterica Retz.:Fr. (Heterobasidiomycetes) Int J Med Mushrooms. 2002;4:215–27.
126. Hsu TH, Lo HC. Biological activity of Cordyceps (Fr.) Link species (ascomycetes) derived from a natural source and from fermented mycelia on diabetes in STZ-induced rats. Int J Med Mushrooms.2002;4:111–25.
127. Kiho T, Ookubo K, Usui S, Ukai S, Hirano K. Structural features and hypoglycaemic activity of a polysaccharide (CS-F10) from the cultured mycelium of Cordyceps sinensis. Biol Pharm Bull. 1999;22:966–70.
128. Mizuno T. Medicinal effects and utilization of Cordyceps (Fr.) Link (Ascomycetes) and Isaria Fr. (mitosporic fungi) Chinese caterpillar fungi, “Tochukaso” (review) Int J Med Mushrooms. 1999;1:251–61.
129. Sato M, Tai T, Nunoura Y, Yajima Y, Kawashima S, Tanaka K. Dehydrotrametenolic acid induces preadipocyte differentiation and sensitizes animal models of noninsulin-dependent diabetes mellitus to insulin. Biol Pharm Bull. 2002;25:81–6.
130. Kim GY, Kim SH, Hwang SY, Kim HY, Park YM, Park SK, et al. Oral administration of proteoglycan isolated from Phellinus linteus in the prevention and treatment of collagen-induced arthritis in mice. Biol Pharm Bull. 2003;26:823–31.
131. Koyama K, Imaizumi T, Akiba M, Kinoshita K, Takahashi L, Suzuki A, et al. Antinociceptive components of Ganoderma lucidum. Planta Med. 1997;63:224–7.
133. Zhang Y, Mills G, Nair MG. Cyclooxygenase inhibitory and antioxidant compounds from the mycelia of the edible mushroom Grifola frondosa. J Agric Food Chem. 2002;50:7581–5.
134. Hirotani M, Ino C, Furuya T, Shiro M. Ganoderic acids T, S and R, new triterpenoids from the cultured mycelia of Ganoderma lucidum. Chem Pharm Bull. 1986;34:2282–5.
135. Chen RY, Yu DQ. Studies on the triterpenoid constituents of the spores from Ganoderma lucidumKarst. J Chin Pharm Sci. 1993;2:91–6.
136. Wang MY, Liu Q, Che QM, Lin ZB. Effects of total triterpenoids extract from Ganoderma lucidum(Curt.:Fr.) P.Karst. (Reishi Mushroom) on experimental liver injury models induced by carbon tetrachloride or d-galactosamine in mice. Int J Med Mushrooms. 2002;4:337–42.
137. Gao Y, Zhou S, Chen G, Dai X, Ye J, Gao H. A phase I/II study of a Ganoderma lucidum (Curt.:Fr.) P.Karst. (Ling Zhi, Reishi mushroom) extract in patients with chronic hepatitis B. Int J Med Mushrooms.2002;4:2321–7.
138. Saito T, Aoki F, Hirai H, Inagaki T, Matsunaga Y, Sakakibara T, et al. Erinacine E as a kappa opioid receptor agonist and its new analogs from a basidiomycete, Hericium ramosum. J Antibiot. 1998;51:983–90.
139. Melzig MF, Pieper S, Siems WE, Heder G, Böttger A, Liberra K, et al. Screening of selected basidiomycetes for inhibitory activity on neutral endopeptidase (NEP) and angiotensin-converting enzyme (ACE) Pharmazie. 1996;51:501–3.
140. Szallasi A, Biro T, Szabo T, Modarres S, Petersen M, Klusch A, et al. A non-pungent triprenyl phenol of fungal origin, scutigeral, stimulates rat dorsal root ganglion neurons via interaction at vanilloid receptors. Br J Pharmacol. 1999;126:1351–8.
141. Liu J. Biologically active substances from mushrooms in Yunnan, China. Heterocycles. 2002;57:157–67.
142. Chang ST. A 40-year journey through bioconversion of lignocellulosic wastes to mushrooms and dietary supplements. Int J Med Mushrooms. 2001;3:299–310.
143. Kim HW, Kim BK. Biomedicinal triterpenoids of Ganoderma lucidum (Curt.: Fr.) P.Karst (Aphyllophoromycetidae) Int J Med Mushrooms. 1999;1:121–38.
144. Mizuno T, Inagaki R, Kanao T, Hagiwara T, Nakamura T, Ito H, et al. Antitumor activity and some properties of water-soluble polysaccharides from “Himematsutake”, the fruiting body of Agaricus blazeiMurill. Agric Biol Chem. 1990;54:2889–96.
145. Mizuno M, Kawakami S, Sakamoto Y, Fujitake N. Macrophages stimulated by polysaccharide purified from Agaricus brasiliensis S.Wasser et al. (Agaricomycetidae) enhance mRNA expression of Th1 cytokine including IL-12 and 18. Int J Med Mushrooms. 2003;5:383–9.
146. Kitamura S, Hori T, Kurita K, Takeo K, Hara C, Itoh W, et al. An antitumor, branched (1-3)-β-d-glucan from a water extract of fruiting bodies of Cryptoporus volvatus. Carbohydr Res. 1994;263:111–21.
147. Watanabe Y, Nakanishi K, Komatsu N, Sakabe T, Terakawa H. Flammulin, an antitumor substance. Bull Chem Soc Jpn. 1964;37:747–50.
148. Lee SS, Lee PL, Chen CF, Wang SY, Chen KY. Antitumor effects of polysaccharides of Ganoderma lucidum (Curt.:Fr.) P. Karst. (Ling Zhi, Reishi Mushroom) (Aphyllophoromycetidae) Int J Med Mushrooms.2003;5:1–16.
149. Ohno N, Adachi Y, Suzuki I, Sato K, Oikawa S, Yadomae T. Characterization of antitumor glucan obtained from liquid-cultured Grifola frondosa. Chem Pharm Bull. 1986;34:1709–15.
150. Chihara G. Medical aspects of lentinan isolated from Lentinus edodes (Berk.) Sing. In: Chang ST, Buswell JA, Chiu SW, editors. Mushroom Biology and Mushroom Products. Hong Kong: The Chinese University Press; 1993. pp. 261–6.
151. Lin Y, Lai P, Huang Y, Xie H. Immune-competent polysaccharides from the submerged cultured mycelium of culinary-medicinal mushroom Lentinus strigellus Berk. & Curt. (Agaricomycetidae) Int J Med Mushrooms. 2004;6:49–55.
152. Han SB, Lee CW, Jeon YJ, Hong ND, Yoo ID, Yang KH, et al. The inhibitory effect of polysaccharides from Phellinus linteus on tumor growth and metastasis. Immunopharmacology.1999;41:157–64.
153. Ohno H, Miura NN, Nakajima M, Yadomae T. Antitumor 1,3-β-glucan from cultured fruit body of Sparassis crispa. Biol Pharmaceut Bull. 2000;23:866–72.
154. Sakagami H, Takeda M. Diverse biological activity of PSK (Krestin), a protein-bound polysaccharide from Coriolus versicolor (Fr.) Quel. In: Chang ST, editor. Mushroom Biology and Mushroom Products.Hong Kong: The Chinese University Press; 1993. pp. 237–45.
155. DeBaets S, Vandamme J. Extracellular Tremella polysaccharides: structures, properties and application. Biotechnol Lett. 2001;23:1361–6.
156. Wang HX, Liu WK, Ng TB, Ooi VEC, Chang ST. Immunomodulatory and antitumor activities of polysaccharide-peptide complex from a mycelial culture of Tricholoma lobayense, a local edible mushroom. Life Sci. 1995;57:269–81.
157. Wang HX, Liu WK, Ng TB, Ooi VEC, Chang ST. The immunomodulatory and antitumor activities of lectins from the mushroom Tricholoma mongolicum. Immunopharmacology. 1996;31:205–11.

Lions Mane: 44 References 1. Mori K, et al Nerve growth factor-inducing activity of Hericium erinaceus in 1321N1 human astrocytoma cells . Biol Pharm Bull. (2008)
Wong KH, et al 2. Neuroregenerative potential of lion’s mane mushroom, Hericium erinaceus (Bull.: Fr.) Pers. (higher Basidiomycetes), in the treatment of peripheral nerve injury (review) . Int J Med Mushrooms. (2012)
3. Bioactive Substances in YAMABUSHITAKE, the Hericium erinaceum Fungus, and its Medicinal Utilization
4. Yaoita Y, Danbara K, Kikuchi M Two new aromatic compounds from Hericium erinaceum (BULL.: FR.) PERS(1) . Chem Pharm Bull (Tokyo). (2005)
5. Chromans, hericenones F, G and H from the mushroom Hericium erinaceum
6. Ueda K, et al An endoplasmic reticulum (ER) stress-suppressive compound and its analogues from the mushroom Hericium erinaceum . Bioorg Med Chem. (2008)
7.Erinacines J and K from the mycelia of Hericium erinaceum
8.Erinacines E, F, and G, stimulators of nerve growth factor (NGF)-synthesis, from the mycelia of Hericium erinaceum
9. Antimicrobial chlorinated orcinol derivatives from mycelia of Hericium erinaceum
10. A sialic acid-binding lectin from the mushroom Hericium erinaceum
11. Li JL, et al A comparative study on sterols of ethanol extract and water extract from Hericium erinaceus . Zhongguo Zhong Yao Za Zhi. (2001)
12. Mizuno T, et al Antitumor-active polysaccharides isolated from the fruiting body of Hericium erinaceum, an edible and medicinal mushroom called yamabushitake or houtou . Biosci Biotechnol Biochem. (1992)
13. Xu H, et al Chemical analysis of Hericium erinaceum polysaccharides and effect of the polysaccharides on derma antioxidant enzymes, MMP-1 and TIMP-1 activities . Int J Biol Macromol. (2010)
14Lee JS, et al Study of macrophage activation and structural characteristics of purified polysaccharides from the fruiting body of Hericium erinaceus . J Microbiol Biotechnol. (2009)
15. Dong Q, Jia LM, Fang JN A beta-D-glucan isolated from the fruiting bodies of Hericium erinaceus and its aqueous conformation . Carbohydr Res. (2006)
16. Han ZH, Ye JM, Wang GF Evaluation of in vivo antioxidant activity of Hericium erinaceus polysaccharides . Int J Biol Macromol. (2013)
17. Abdullah N, et al Evaluation of Selected Culinary-Medicinal Mushrooms for Antioxidant and ACE Inhibitory Activities . Evid Based Complement Alternat Med. (2012)
18. Effects of cultivation techniques and processing on antimicrobial and antioxidant activities of Hericium erinaceus ( Bull .: Fr .) Pers . Extracts
19. Erinacines A, B and C, strong stimulators of nerve growth factor (NGF)-synthesis, from the mycelia of Hericium erinaceum
20. Neurotropic and Trophic Action of Lion’s Mane Mushroom Hericium erinaceus (Bull.: Fr.) Pers. (Aphyllophoromycetideae) Extracts on Nerve Cells in Vitro
21. Ueda K, et al Endoplasmic reticulum (ER) stress-suppressive compounds from scrap cultivation beds of the mushroom Hericium erinaceum . Biosci Biotechnol Biochem. (2009)
22. Kolotushkina EV, et al The influence of Hericium erinaceus extract on myelination process in vitro . Fiziol Zh. (2003)
23. Mori K, et al Effects of Hericium erinaceus on amyloid β(25-35) peptide-induced learning and memory deficits in mice . Biomed Res. (2011)
24. Mori K, et al Improving effects of the mushroom Yamabushitake (Hericium erinaceus) on mild cognitive impairment: a double-blind placebo-controlled clinical trial . Phytother Res. (2009)
25. Nagano M, et al Reduction of depression and anxiety by 4 weeks Hericium erinaceus intake . Biomed Res. (2010)
26. Wong KH, et al Peripheral Nerve Regeneration Following Crush Injury to Rat Peroneal Nerve by Aqueous Extract of Medicinal Mushroom Hericium erinaceus (Bull.: Fr) Pers. (Aphyllophoromycetideae) . Evid Based Complement Alternat Med. (2011)
27. Activity of Aqueous Extracts of Lion’s Mane Mushroom Hericium erinaceus (Bull.: Fr.) Pers. (Aphyllophoromycetideae) on the Neural Cell Line NG108-15
28. Farndale RW, et al The role of collagen in thrombosis and hemostasis . J Thromb Haemost. (2004)
29. Mori K, et al Inhibitory effect of hericenone B from Hericium erinaceus on collagen-induced platelet aggregation . Phytomedicine. (2010)
30. Angiotensin I-converting enzyme inhibitor from Grifola frondosa
31. Isolation and characterization of a novel angiotensin I-converting enzyme inhibitory peptide derived from the edible mushroom Tricholoma giganteum
32. Antioxidative and ACE inhibitory activities in enzymatic hydrolysates of the cotton leafworm, Spodoptera littoralis
33. Hiwatashi K, et al Yamabushitake mushroom (Hericium erinaceus) improved lipid metabolism in mice fed a high-fat diet . Biosci Biotechnol Biochem. (2010)
34. Yang BK, Park JB, Song CH Hypolipidemic effect of an Exo-biopolymer produced from a submerged mycelial culture of Hericium erinaceus . Biosci Biotechnol Biochem. (2003)
35. Kim YO, et al Hericium erinaceus suppresses LPS-induced pro-inflammation gene activation in RAW264.7 macrophages . Immunopharmacol Immunotoxicol. (2011)
36. Wang JC, et al Antitumor and immunoenhancing activities of polysaccharide from culture broth of Hericium spp . Kaohsiung J Med Sci. (2001)
37. Abdulla MA, et al Potential activity of aqueous extract of culinary-medicinal Lion’s Mane mushroom, Hericium erinaceus (Bull.: Fr.) Pers. (Aphyllophoromycetideae) in accelerating wound healing in rats . Int J Med Mushrooms. (2011)
38. Kim SP, Nam SH, Friedman M Hericium erinaceus (Lion’s Mane) mushroom extracts inhibit metastasis of cancer cells to the lung in CT-26 colon cancer-tansplanted mice . J Agric Food Chem. (2013)
39. Park ID, et al Toxicological study on MUNOPHIL, water extract of Panax ginseng and Hericium erinaceum in rats . J Acupunct Meridian Stud. (2008)
40. Nakatsugawa M, et al Hericium erinaceum (yamabushitake) extract-induced acute respiratory distress syndrome monitored by serum surfactant proteins . Intern Med. (2003)
41. A method for estimating the probability of adverse drug reactions
42. Tanaka A, Matsuda H Expression of nerve growth factor in itchy skins of atopic NC/NgaTnd mice . J Vet Med Sci. (2005)
43. Mori K, et al Improving effects of the mushroom Yamabushitake (Hericium erinaceus) on mild cognitive impairment: a double-blind placebo-controlled clinical trial . Phytother Res. (2009)
44. Nagano M, et al Reduction of depression and anxiety by 4 weeks Hericium erinaceus intake . Biomed Res. (2010)

Oyster Var. : 41 References 1. Effects of edible mushrooms (Pleurotus ostreatus (Jacq.) P. Kumm., Pleurotus eryngil, Flammulina velutipes) extracts on immune cell activation in mice (830.17) 2. T. C. Finimundy, G. Gambato, R. Fontana et al., “Aqueous extracts of Lentinula edodes and Pleurotus sajor-caju exhibit high antioxidant capability and promising in vitro antitumor activity,” Nutrition Research, vol. 33, no. 1, pp. 76–84, 2013.
3. A. Synytsya, K. Míčková, I. Jablonský et al., “Glucans from fruit bodies of cultivated mushrooms Pleurotus ostreatus and Pleurotus eryngii: structure and potential prebiotic activity,” Carbohydrate Polymers, vol. 76, no. 4, pp. 548–556, 2009. 4. J.-C. Wang, S.-H. Hu, Z.-C. Liang, and C.-J. Yeh, “Optimization for the production of water-soluble polysaccharide from Pleurotus citrinopileatus in submerged culture and its antitumor effect,” Applied Microbiology and Biotechnology, vol. 67, no. 6, pp. 759–766, 2005. 5. T. Jayakumar, M. Sakthivel, P. A. Thomas, and P. Geraldine, “Pleurotus ostreatus, an oyster mushroom, decreases the oxidative stress induced by carbon tetrachloride in rat kidneys, heart and brain,” Chemico-Biological Interactions, vol. 176, no. 2-3, pp. 108–120, 2008.
6. A. Jedinak and D. Sliva, “Pleurotus ostreatus inhibits proliferation of human breast and colon cancer cells through p53-dependent as well as p53-independent pathway,” International Journal of Oncology, vol. 33, no. 6, pp. 1307–1313, 2008.
7. I. Lavi, D. Friesem, S. Geresh, Y. Hadar, and B. Schwartz, “An aqueous polysaccharide extract from the edible mushroom Pleurotus ostreatus induces anti-proliferative and pro-apoptotic effects on HT-29 colon cancer cells,” Cancer Letters, vol. 244, no. 1, pp. 61–70, 2006.
8. K. K. Mishra, R. S. Pal, R. Arunkumar, C. Chandrashekara, S. K. Jain, and J. C. Bhatt, “Antioxidant properties of different edible mushroom species and increased bioconversion efficiency of Pleurotus eryngii using locally available casing materials,” Food Chemistry, vol. 138, no. 2-3, pp. 1557–1563, 2013.
9. K. Mori, C. Kobayashi, T. Tomita, S. Inatomi, and M. Ikeda, “Antiatherosclerotic effect of the edible mushrooms Pleurotus eryngii (Eringi), Grifola frondosa (Maitake), and Hypsizygus marmoreus (Bunashimeji) in apolipoprotein E-deficient mice,” Nutrition Research, vol. 28, no. 5, pp. 335–342, 2008.
10. L. K. Jagadish, R. Shenbhagaraman, V. Venkatakrishnan, and V. Kaviyarasan, “Studies on the phytochemical, antioxidant and antimicrobial properties of three indigenous Pleurotus species,” Journal of Molecular Biology and Biotechnology, vol. 1, pp. 20–29, 2008.
11. A. Jedinak, S. Dudhgaonkar, Q.-L. Wu, J. Simon, and D. Sliva, “Anti-inflammatory activity of edible oyster mushroom is mediated through the inhibition of NF-κB and AP-1 signaling,” Nutrition Journal, vol. 10, article 52, 2011.
12. G. Kanagasabapathy, S. N. A. Malek, U. R. Kuppusamy, and S. Vikineswary, “Chemical composition and antioxidant properties of extracts of fresh fruiting bodies of Pleurotus sajor-caju (Fr.) singer,” Journal of Agricultural and Food Chemistry, vol. 59, no. 6, pp. 2618–2626, 2011 13. K. K. Maity, S. Patra, B. Dey et al., “A heteropolysaccharide from aqueous extract of an edible mushroom, Pleurotus ostreatus cultivar: structural and biological studies,” Carbohydrate Research, vol. 346, no. 2, pp. 366–372, 2011. 14. H. Tong, F. Xia, K. Feng et al., “Structural characterization and in vitro antitumor activity of a novel polysaccharide isolated from the fruiting bodies of Pleurotus ostreatus,” Bioresource Technology, vol. 100, no. 4, pp. 1682–1686, 2009. 15. P. Bobek, L. Ozdín, and Š. Galbavý, “Dose- and time-dependent hypocholesterolemic effect of oyster mushroom (Pleurotus ostreatus) in rats,” Nutrition, vol. 14, no. 3, pp. 282–286, 1998. 16. P. H. K. Ngai and T. B. Ng, “A ribonuclease with antimicrobial, antimitogenic and antiproliferative activities from the edible mushroom Pleurotus sajor-caju,” Peptides, vol. 25, no. 1, pp. 11–17, 2004. 17. S.-M. Wong, K.-K. Wong, L. C.-M. Chiu, and P. C.-K. Cheung, “Non-starch polysaccharides from different developmental stages of Pleurotus tuber-regium inhibited the growth of human acute promyelocytic leukemia HL-60 cells by cell-cycle arrest and/or apoptotic induction,” Carbohydrate Polymers, vol. 68, no. 2, pp. 206–217, 2007. 18. Y. R. Li, Q. H. Liu, H. X. Wang, and T. B. Ng, “A novel lectin with potent antitumor, mitogenic and HIV-1 reverse transcriptase inhibitory activities from the edible mushroom Pleurotus citrinopileatus,” Biochimica et Biophysica Acta, vol. 1780, no. 1, pp. 51–57, 2008.
19. C.-W. Phan, W.-L. Wong, P. David, M. Naidu, and V. Sabaratnam, “Pleurotus giganteus (Berk.) Karunarathna & K.D. Hyde: nutritional value and in vitro neurite outgrowth activity in rat pheochromocytoma cells,” BMC Complementary and Alternative Medicine, vol. 12, article 102, 2012. 20. W.-L. Wong, M. A. Abdulla, K.-H. Chua, U. R. Kuppusamy, Y.-S. Tan, and V. Sabaratnam, “Hepatoprotective effects of Panus giganteus (Berk.) corner against thioacetamide-(TAA-) induced liver injury in rats,” Evidence-Based Complementary and Alternative Medicine, vol. 2012, Article ID 170303, 10 pages, 2012. 21. Gunde-Cimerman NG: Medicinal value of the genus Pleurotus (Fr.) P. Karst. (Agricales S.R., Basidiomycetes). International Journal of Medicinal mushrooms. 1999 22. Bobek P, Galbavy S: Hypocholesterolemic and antiatherogenic effect of oyster mushroom (Pleurotus ostreatus) in rabbits. Nahrung. 1999 23. Bobek P, Ozdin L, Galbavy S: Dose and time-dependent hypocholesterolemic effect of oyster mushroom (Pleurotus ostreatus) in rats. Nutrition. 1998 24. Bobek P, Ozdin L, Kuniak L: Mechanism of hypocholesterolemic effect of oyster mushroom (Pleurotus ostreatus) in rats: reduction of cholesterol absorption and increase of plasma cholesterol removal Z. Ernahrungswiss. 1994,
25. Antihyperlipidemic effects of Pleurotus ostreatus (oyster mushrooms) in HIV-infected individuals taking antiretroviral therapy, Donald I Abrams. 2011.
26. Anti-inflammatory activity of edible oyster mushroom is mediated through the inhibition of NF-κB and AP-1 signaling. Andrej Jedinak†, Shailesh Dudhgaonkar†, Qing-li Wu, James Simon and Daniel Sliva. 2011. 27. Hagiwara SY, et al A phytochemical in the edible Tamogi-take mushroom (Pleurotus cornucopiae), D-mannitol, inhibits ACE activity and lowers the blood pressure of spontaneously hypertensive rats . Biosci Biotechnol Biochem. (2005)
33. Inhibitory effects of l-pipecolic acid from the edible mushroom, Sarcodon aspratus, on angiotensin I-converting enzyme 34. Jose N, Ajith TA, Jananrdhanan KK. Antioxidant, anti-inflammatory, and antitumor activities of culinary-medicinal mushroom Pleurotus pulmonarius (Fr.) Quél. (Agaricomycetidae) Int J Med Mushrooms.2002;4:329–35.
35. Bobek P, Galbavy` Š, Ozdiĭn L. Effect of oyster mushroom (Pleurotus ostreatus) on pathological changes in dimethylhydrazine-induced rat colon cancer. Oncol Rep. 1998;5:727–30. 36. Wang HX, Gao J, Ng TB. A new lectin with highly potent antihepatoma and antisarcoma activities from the oyster mushroom Pleurotus ostreatus. Biochem Biophys Res Commun. 2000;275:810–6. 37. Zusman I, Reifen R, Livni O, Smirnoff P, Gurevich P, Sandler B, et al. Role of apoptosis, proliferating cell nuclear antigen and p53 protein in chemically induced colon cancer in rats fed corncob fiber treated with the fungus Pleurotus ostreatus. Anticancer Res. 1997;17:2105–13. 38. Gerasimenya VP, Efremenkova OV, Kamzolina OV, Bogush TA, Tolstych IV, Zennkova VA. Antimicrobial and antitoxical action of edible and medicinal mushroom Pleurotus ostreatus (Jacq,:Fr.) Kumm. Extracts. Int J Med Mushrooms. 2002;4:127–32. 39. Lakshmi B, Jose N, Ajith TA, Jananrdhanan KK. Antimutagenic activity of methanolic extract of culinary-medicinal oyster mushroom, Pleurotus ostreatus (Jacq.:Fr.) Kumm. (strain floric Eger nom. Nud.) and its protective effect against benzo[a]pyrene-induced hepatic damages. Int J Med Mushrooms.2004;6:139–49. 40. Bobek P, Galbavy` Š. Hypocholesteremic and antiatherogenic effect of oyster mushroom (Pleurotus ostreatus) in rabbits. Nahrung. 1999;43:339–42.
41. Gunde-Cimerman N, Friedrich J, Cimerman A, Benički N. Screening fungi for the production of an inhibitor of HMG-CoA reductase—production of mevinolin by the fungi of the genus Pleurotus. FEMS Microbiol Lett. 1993;111:203–6.