top of page

Medicinal mushrooms are known for their nutritional and healthful value and for the diversity of the bioactive compounds they contain. Mushrooms have a cross-cultural history of medicinal use spanning many millennia. In Asia, South America, Africa and throughout Europe, mushrooms have been used medicinally and as food, while others have been used in rituals to awaken consciousness. Fungi and mushrooms are extremely abundant worldwide and show diversity. The number of mushroom species on earth is currently estimated at 150,000-160,000, yet maybe only 10% are known.  

 

Medicinal mushrooms comprise a vast and yet largely untapped source of powerful new chemical and pharmaceutical products. They represent an unlimited source of nutrients and compounds with antitumor and immuno-stimulating properties. The chief medicinal uses of mushrooms discovered so far are as antitumor, immunomodulating, antioxidant, radical scavenging, cardiovascular, antiviral, antibacterial, antiparasitic, antifungal, detoxicating, hepatoprotective, and antidiabetic effects. The best usage of medicinal mushrooms has been in preventing and treating immune disorders, especially in immunodeficient and immunosuppressed patients. They are also used for patients under chemotherapy or radiotherapy; in different types of cancers, chronic bloodborne viral infections of Hepatitis B, C, and D, different types of anemia, the human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS), Herpes simplex virus (HSV), chronic fatigue syndrome, Epstein-Barr virus; for patients with chronic gastritis and gastric ulcers caused by Helicobacter pylori; and for patients with dementia (especially for Alzheimer’s disease).

 

In traditional Chinese and Ayurvedic medicine, mushrooms’ legendary effects on promoting health and vitality and in fighting tumors have been extensively validated in numerous studies. Mushrooms have also played an important role in the treatment of ailments affecting the rural populations of eastern European countries. Modern clinical practice in Japan, China, Korea, Russia, and several other countries also utilize mushroom-derived preparations. There is also a long history of traditional use of mushrooms as curatives in Mesoamerica (especially for species of the genus Psilocybe), Africa (Yoruba populations in Nigeria and Benin), Algeria, and Egypt. A very special role of Amanita muscaria is found in Siberia and Tibetan shamanism, Buddhism, and Celtic myths.

 

Studies suggest that specific mushrooms are strongly immunologic and can help us maintain physiological homeostasis, restore physical balance, and improve our natural resistance to disease. More than 270 recognized species of mushrooms are known to have specific immunotherapeutic properties. Numerous studies have demonstrated that certain components present in medicinal mushrooms have been responsible for the modulation of cellular and physiological changes in the host. It is for this reason that mushrooms are often used in numerous illnesses, in part as cancer therapeutic agents. In fact, medicinal mushrooms probably hold one of the greatest immunological promises for the future of oncology treatment. 

 

Mushrooms contain unique chemicals: polysaccharides, lectins, lipids, hericenone, 
erinacol, erinacine, terpenoids and numerous others. Mushroom polysaccharides in particular prevent oncogenesis, show direct antitumor activity against various allogeneic and syngenetic tumors, and prevent tumor metastasis. Polysaccharides from mushrooms do not attack cancer cells directly, but produce antitumor effects by activating different immune responses in the host. These substances are regarded as biological response modifiers. This basically means that: (1) they cause no harm and place no additional stress on the body; (2) they help the body adapt to various environmental and biological stresses; and (3) they exert a non-specific action on the body, supporting some or all of the major systems, including nervous, hormonal, and immune systems, as well as regulatory functions. 

 

The mushrooms credited with success against cancer and various diseases belong to the genus Phellinus, Pleurotus, Agaricus, 
Ganoderma, Clitocybe, Antrodia, Trametes, 
Cordyceps, Xerocomus, Calvatia, 
Schizophyllum, Flammulina, Suillus, Inonotus, 
Inocybe, Funlia, Lactarius, Albatrellus, Russula,
and Fomes. The anticancer compounds play a crucial role as reactive oxygen species inducer, mitotic kinase inhibitor, anti-mitotic, angiogenesis inhibitor, and topoisomerase inhibitor, leading to apoptosis, and eventually checking cancer proliferation.

 

Studies show medicinal mushrooms exhibit no chronic or acute toxicity. Cell nucleus studies show no detrimental effects and DNA has shown no mutations. Pregnant animal studies have demonstrated there is no detriment to fetal development, and no LD50 (a measure of toxicity). Medicinal mushrooms apparently produce no harmful side effects.

 

The following are selected articles on medicinal mushrooms. An attempt has been made to categorize articles by specific medicinal mushroom types: Agaricus blazei, Cordyceps sinensis, Coriolus versicolor, Grifola frondosa, Ganoderma lucidum, Hericium erinaceus, Inonotus obliquus, and Lentinus edodes

Aghili, S. R., T. Shokohi, G. H. Janbabai, M. Karimi, N. Aslani, and B. Salmanian. Anti-tumor Compounds of Higher Basidiomycetes Fungi: A Review of Novel Known Mushroom Derivatives with Anti-tumor Function. Journal of Medicinal Plants 2, no. 58 (2016): 7-24. (Article in Arabic) 

Aida, F. M. N. A., M. Shuhaimi, M. Yazid, and A. G. Maaruf. Mushroom as a potential source of prebiotics: a review. Trends in Food Science & Technology 20, no. 11-12 (2009): 567-575.

 

Ajith, Thekkuttuparambil A., and Kainoor K. Janardhanan. Indian medicinal mushrooms as a source of antioxidant and antitumor agents. Journal of Clinical Biochemistry and Nutrition 40, no. 3 (2007): 157-162.

Alves, Maria José, Isabel CFR Ferreira, Joana Dias, Vânia Teixeira, Anabela Martins, and Manuela Pintado. A review on antimicrobial activity of mushroom (Basidiomycetes) extracts and isolated compounds. Planta medica 78, no. 16 (2012): 1707-1718.

 

Asatiani, Mikheil D., Vladimir Elisashvili, George Songulashvili, Abraham Z. Reznick, and Solomon P. Wasser. Higher basidiomycetes mushrooms as a source of antioxidants. In Progress in Mycology, pp. 311-326. Springer, Dordrecht, 2010.

 

Borchers, A. T.; Krishnamurthy, A; Keen, C. L.; Meyers, F. J.; Gershwin, M. E. (2008). The immunobiology of mushrooms.  Experimental Biology and Medicine 233 (3): 259–76.

Borchers, Andrea T., Carl L. Keen, and M. Eric Gershwin. Mushrooms, tumors, and immunity: an update. Experimental Biology and Medicine 229, no. 5 (2004): 393-406.

 

Ferreira, Isabel CFR, Lillian Barros, and Rui Abreu. Antioxidants in wild mushrooms. Current Medicinal Chemistry 16, no. 12 (2009): 1543-1560.

 

Ferreira, Isabel CFR, Josiana A Vaz, M. Helena Vasconcelos, and Anabela Martins. Compounds from wild mushrooms with antitumor potential. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents) 10, no. 5 (2010): 424-436.

 

Ganeshpurkar, Aditya, Gopal Rai, and Alok Pal Jain. Medicinal mushrooms: Towards a new horizon. Pharmacognosy reviews 4, no. 8 (2010): 127.

 

Guggenheim, Alena G., Kirsten M. Wright, and Heather L. Zwickey. Immune modulation from five major mushrooms: application to integrative oncology. Integrative Medicine: A Clinician's Journal 13, no. 1 (2014): 32.

 

Kidd, Parris M. The use of mushroom glucans and proteoglycans in cancer treatment. Alternative medicine review 5, no. 1 (2000): 4-27.

 

Lindequist, Ulrike, Ha Won Kim, Evelin Tiralongo, and Leo Van Griensven. Medicinal mushrooms. Evidence-Based Complementary and Alternative Medicine 2014 (2014).

 

Lindequist, Ulrike, Timo HJ Niedermeyer, and Wolf-Dieter Jülich. The pharmacological potential of mushrooms. Evidence-Based Complementary and Alternative Medicine 2, no. 3 (2005): 285-299.

 

Monro, Jean (2003). Treatment of Cancer with Mushroom Products. Archives of Environmental Health: an International Journal 58 (8): 533-7. 

 

Ooi VE, Liu F. Immunomodulation and anti-cancer activity of polysaccharide-protein complexes. Curr Med Chem. 2000; 7:715–729. 

 

Poucheret, Patrick, Francoise Fons, and Sylvie Rapior. Biological and pharmacological activity of higher fungi: 20-year retrospective analysis. Cryptogamie Mycologie 27, no. 4 (2006): 311.

 

Smith, John E., Neil J. Rowan, and Richard Sullivan. Medicinal mushrooms: a rapidly developing area of biotechnology for cancer therapy and other bioactivities. Biotechnology Letters 24, no. 22 (2002): 1839-1845.

 

Stamets, Paul, and Heather Zwickey. Medicinal mushrooms: ancient remedies meet modern science. Integrative Medicine: A Clinician's Journal 13, no. 1 (2014): 46.

 

Wani, Bilal Ahmad, R. H. Bodha, and A. H. Wani. Nutritional and medicinal importance of mushrooms. Journal of Medicinal Plants Research 4, no. 24 (2010): 2598-2604.

 

Wasser, Solomon. Medicinal mushroom science: Current perspectives, advances, evidences, and challenges. Biomedical journal 37, no. 6 (2014).

 

Wasser, Solomon P., and Alexander L. Weis. Medicinal properties of substances occurring in higher basidiomycetes mushrooms: current perspectives. International Journal of medicinal mushrooms 1, no. 1 (1999).

 

Zaidman, Ben-Zion, Majed Yassin, Jamal Mahajna, and Solomon P. Wasser. Medicinal mushroom modulators of molecular targets as cancer therapeutics. Applied Microbiology and Biotechnology 67, no. 4 (2005): 453-468.

Agaricus bisporus White Button Mushroom

 

Adams, Lynn S., Shiuan Chen, Sheryl Phung, Xiwei Wu, and Lui Ki. White button mushroom (Agaricus bisporus) exhibits antiproliferative and proapoptotic properties and inhibits prostate tumor growth in athymic mice. Nutrition and cancer 60, no. 6 (2008): 744-756.

 

Bonnen, Alice M., Lori H. Anton, and Ann B. Orth. Lignin-degrading enzymes of the commercial button mushroom, Agaricus bisporus. Appl. Environ. Microbiol. 60, no. 3 (1994): 960-965.

 

Chen, Shiuan, Sei-Ryang Oh, Sheryl Phung, Gene Hur, Jing Jing Ye, Sum Ling Kwok, Gayle E. Shrode, Martha Belury, Lynn S. Adams, and Dudley Williams. Anti-aromatase activity of phytochemicals in white button mushrooms (Agaricus bisporus). Cancer research 66, no. 24 (2006): 12026-12034.

 

Dhamodharan, G., and S. Mirunalini. A Novel Medicinal Characterization of Agaricus bisporus (white button mushroom). Pharmacology online 2 (2010): 456-463.

 

Grube, Baiba J., Elizabeth T. Eng, Yeh-Chih Kao, Annette Kwon, and Shiuan Chen. White button mushroom phytochemicals inhibit aromatase activity and breast cancer cell proliferation. The Journal of nutrition 131, no. 12 (2001): 3288-3293.

 

Hong, Seo Ah, Kirang Kim, Seok‐Jin Nam, Gu Kong, and Mi Kyung Kim. A case–control study on the dietary intake of mushrooms and breast cancer risk among Korean women. International journal of cancer 122, no. 4 (2008): 919-923.

 

Jeong, Sang Chul, Yong Tae Jeong, Byung Keun Yang, Rezuanul Islam, Sundar Rao Koyyalamudi, Gerald Pang, Kai Yip Cho, and Chi Hyun Song. White button mushroom (Agaricus bisporus) lowers blood glucose and cholesterol levels in diabetic and hypercholesterolemic rats. Nutrition research 30, no. 1 (2010): 49-56.

 

Kanaya, Noriko, Makoto Kubo, Zheng Liu, Peiguo Chu, Charles Wang, Yate-Ching Yuan, and Shiuan Chen. Protective effects of white button mushroom (Agaricus bisporus) against hepatic steatosis in ovariectomized mice as a model of postmenopausal women. PLoS One 6, no. 10 (2011): e26654.

 

Ren, Zhihong, Zhuyan Guo, Simin Nikbin Meydani, and Dayong Wu. White button mushroom enhances maturation of bone marrow-derived dendritic cells and their antigen presenting function in mice. The Journal of nutrition 138, no. 3 (2008): 544-550.

 

Savoie, Jean‐Michel, Nathalie Minvielle, and Michèle L. Largeteau. Radical‐scavenging properties of extracts from the white button mushroom, Agaricus bisporus. Journal of the Science of Food and Agriculture 88, no. 6 (2008): 970-975.

 

Soković, Marina, and Leo JLD van Griensven. Antimicrobial activity of essential oils and their components against the three major pathogens of the cultivated button mushroom, Agaricus bisporus. European Journal of Plant Pathology 116, no. 3 (2006): 211-224.

 

Wang, Junpeng, Xinli Niu, Xiaogang Du, Donald Smith, Simin Nikbin Meydani, and Dayong Wu. Dietary supplementation with white button mushrooms augments the protective immune response to Salmonella vaccine in mice. The Journal of nutrition 144, no. 1 (2013): 98-105.

 

Wu, Dayong, Munkyong Pae, Zhihong Ren, Zhuyan Guo, Donald Smith, and Simin Nikbin Meydani. Dietary supplementation with white button mushroom enhances natural killer cell activity in C57BL/6 mice. The Journal of nutrition 137, no. 6 (2007): 1472-1477.

 

Zhang, Min, Jian Huang, Xing Xie, and C. D'Arcy J. Holman. Dietary intakes of mushrooms and green tea combine to reduce the risk of breast cancer in Chinese women. International journal of cancer 124, no. 6 (2009): 1404-1408.

 

Agaricus blazei Murill

 

Ahn WS, Kim DJ, Chae GT, Lee JM, Bae SM, Sin JI, Kim YW, Namkoong SE, Lee IP. Natural killer cell activity and quality of life were improved by consumption of a mushroom extract, Agaricus blazei Murill Kyowa, in gynecological cancer patients undergoing chemotherapy. International Journal of Gynecological Cancer 2004;14(4):589-94.

 

Akiyama H, Endo M, Matsui T, et al. Agaritine from Agaricus blazei Murrill induces apoptosis in the leukemic cell line U937. Biochim Biophysica Acta. May 2011;1810(5):519-525.

 

Endo M, Beppu H, Akiyama H et al., Agaritine purified from Agaricus blazei Murrill exerts anti-tumor activity against leukemic cells. Biochimica et Biophysica Acta, vol. 1800, no. 7, pp. 669–673, 2010.

 

Ishii PL, Prado CK, de Mauro MO, et al. Evaluation of Agaricus blazei in vivo for antigenotoxic, anticarcinogenic, phagocytic and immunomodulatory activities. Regul Toxicol Pharmacol. 2011;59(3):412–422.

 

Kimura Y, Kido T, Takaku T, et al. Isolation of an anti-angiogenic substance from Agaricus blazei Murill: its antitumor and antimetastatic actions. Cancer Sci. Sep 2004;95(9):758-764.

 

Lee YL, Kim HJ, Lee MS, et al. Oral administration of Agaricus blazei (H1 strain) inhibited tumor growth in a sarcoma 180 inoculation model. Exp Anim. Oct 2003;52(5):371-375.

 

Lin JG, Fan MJ, Tang NY, et al. An extract of Agaricus blazei Murill administered orally promotes immune responses in murine leukemia BALB/c mice in vivo. Integr Cancer Ther. 2012;11(1):29–36.

 

Mizuno M, Morimoto M, Minato K-I, Tsuchida H. Polysaccharides from Agaricus blazei stimulate lymphocyte T-cell subsets in mice. Bioscience, Biotechnology and Biochemistry.1998;62(3):434–437. 

 

Murakawa K, Fukunaga K, Tanouchi M, Hosokawa M, Hossain Z, and Takahashi K. Therapy of myeloma in vivo using marine phospholipid in combination with Agaricus blazei Murill as an immune respond activator. Journal of Oleo Science, vol. 56, no. 4, pp. 179–188, 2007.

 

Ohno S, Sumiyoshi Y, Hashine K, Shirato A, Kyo S, Inoue M. Phase I clinical study of the dietary supplement, Agaricus blazei Murill, in cancer patients in remission. Evid Based Complement Alternat Med. 2011;2011:192381.

 

Sorimachi K, Akimoto K, Ikehara Y, Inafuku K, Okubo A, and Yamazaki S. Secretion of TNF-α, IL-8 and nitric oxide by macrophages activated with Agaricus blazei Murill fractions in vitro. Cell Structure and Function, vol. 26, no. 2, pp. 103–108, 2001.

 

Takaku T, Kimura Y, and Okuda H. Isolation of an antitumor compound from Agaricus blazei Murill and its mechanism of action. Journal of Nutrition, vol. 131, no. 5, pp. 1409–1413, 2001.

 

Takimoto H, Wakita D, Kawaguchi K, et al. Potentiation of cytotoxic activity in naive and tumor-bearing mice by oral administration of hot-water extracts from Agaricus blazei fruiting bodies. Biol Pharm Bull. Mar 2004;27(3):404-406.

 

Yuminamochi E, Koike T, Takeda K, et al. Interleukin-12- and interferon-gamma-mediated natural killer cell activation by Agaricus blazei Murill. Immunology. Jun 2007;121(2):197-206.

 

 

Cordyceps sinensis “Dong Chong Xia Cao”

 

Bok, Jin Woo, Leonard Lermer, Jeff Chilton, Hans G. Klingeman, and GH Neil Towers. Antitumor sterols from the mycelia of Cordyceps sinensis. Phytochemistry 51, no. 7 (1999): 891-898.

 

Chen, Y., H. Guo, Z. Du, X‐Z. Liu, Y. Che, and X. Ye. Ecology‐based screen identifies new metabolites from a Cordyceps‐colonizing fungus as cancer cell proliferation inhibitors and apoptosis inducers. Cell Proliferation 42, no. 6 (2009): 838-847.

 

Das, Shonkor Kumar, Mina Masuda, Akihiko Sakurai, and Mikio Sakakibara. Medicinal uses of the mushroom Cordyceps militaris: current state and prospects. Fitoterapia 81, no. 8 (2010): 961-968.

 

Jayakumar, Thanasekaran, Chong-Chi Chiu, Shwu-Huey Wang, Duen-Suey Chou, Yung-Kai Huang, and Joen-Rong Sheu. Anti-cancer effects of CME-1, a novel polysaccharide, purified from the mycelia of Cordyceps sinensis against B16-F10 melanoma cells. Journal of cancer research and therapeutics 10, no. 1 (2014): 43.

 

Jeong, Min-Ho, Chang-Min Lee, Sang-Wha Lee, Su-Yeong Seo, Min-Jeong Seo, Byoung-Won Kang, Yong-Kee Jeong, Yoo-Jin Choi, Kwang-Mo Yang, and Wol-Soon Jo. Cordycepin-enriched Cordyceps militaris induces immunomodulation and tumor growth delay in mouse-derived breast cancer. Oncology reports 30, no. 4 (2013): 1996-2002.

 

Ji NF, Yao LS, Li Y, He W, Yi KS, Huang M. Polysaccharide of Cordyceps sinensis enhances cisplatin cytotoxicity in non-small cell lung cancer H157 cell line. Integr Cancer Ther. 2011;10(4):359–367.

 

Koh, Jong-Ho, Jin-Man Kim, Un-Jae Chang, and Hyung-Joo Suh. Hypocholesterolemic effect of hot-water extract from mycelia of Cordyceps sinensis. Biological and Pharmaceutical Bulletin 26, no. 1 (2003): 84-87.

 

Li, S. P., Ping Li, T. T. X. Dong, and K. W. K. Tsim. Anti-oxidation activity of different types of natural Cordyceps sinensis and cultured Cordyceps mycelia. Phytomedicine 8, no. 3 (2001): 207-212.

 

Niwa Y, Matsuura H, Murakami M, Sato J, Hirai K, Sumi H. Evidence that naturopathic therapy including Cordyceps sinensis prolongs survival of patients with hepatocellular carcinoma. Integr Cancer Ther. 2013;12(1):50–68. 

 

Paterson, R. Russell M. Cordyceps–a traditional Chinese medicine and another fungal therapeutic biofactory? Phytochemistry 69, no. 7 (2008): 1469-1495.

 

Won, So-Young, and Eun-Hee Park. Anti-inflammatory and related pharmacological activities of cultured mycelia and fruiting bodies of Cordyceps militaris. Journal of ethnopharmacology 96, no. 3 (2005): 555-561.

 

Zhu, Jia-Shi, Georges M. Halpern, and Kenneth Jones. The scientific rediscovery of a precious ancient Chinese herbal regimen: Cordyceps sinensis Part II. The Journal of Alternative and Complementary Medicine 4, no. 4 (1998): 429-457.

 

 

Coriolus versicolor aka Trametes versicolor 
Yun Zhi – 
polysaccharide-K (PSK) and polysaccharopeptide (PSP)

 

Adachi K, Nanba H, Kuroda H. Potentiation of host-mediated antitumor activity in mice by beta glucan obtained from Grifola frondosa (maitake). Chem Pharm Bull 1987; 35:262-70.

Chang, Yajing, Meng Zhang, Yifei Jiang, Yong Liu, Heng Luo, Cui Hao, Pengjiao Zeng, and Lijuan Zhang. "Preclinical and clinical studies of Coriolus versicolor polysaccharopeptide as an immunotherapeutic in China." Discovery Medicine 23, no. 127 (2017): 207-219.


Chay, Wen Yee, Chee Kian Tham, Han Chong Toh, Hwee Yong Lim, Chee Kiat Tan, Cindy Lim, Who-Whong Wang, and Su-Pin Choo. "Coriolus versicolor (Yunzhi) use as therapy in advanced hepatocellular carcinoma patients with poor liver function or who are unfit for standard therapy." The Journal of Alternative and Complementary Medicine 23, no. 8 (2017): 648-652.

 

Chu, Kevin KW, Susan SS Ho, and Albert HL Chow. Coriolus versicolor: a medicinal mushroom with promising immunotherapeutic values. The Journal of Clinical Pharmacology 42, no. 9 (2002): 976-984.

 

CÓRDOBA M, Ketty A., and Alicia RÍOS H. "Biotechnological applications and potential uses of the mushroom Tramestes versicolor." Vitae 19, no. 1 (2012): 70-76.

Cruz, Aritson, Lígia Pimentel, Luis M. Rodríguez-Alcalá, Tito Fernandes, and Manuela Pintado. "Health benefits of edible mushrooms focused on Coriolus versicolor: A review." Journal of Food and Nutrition Research 4, no. 12 (2016): 773-781.

chrome-

 

Cui, Jian, and Yusuf Chisti. Polysaccharopeptides of Coriolus versicolor: physiological activity, uses, and production. Biotechnology advances 21, no. 2 (2003): 109-122.

 

Dou, Huaiqian, Yajing Chang, and Lijuan Zhang. "Coriolus versicolor polysaccharopeptide as an immunotherapeutic in China." Progress in Molecular Biology and Translational Science 163 (2019): 361-381.

 

LY Eliza, Wong, Cheng K Fai, and Leung P Chung. "Efficacy of Yun Zhi (Coriolus versicolor) on survival in cancer patients: systematic review and meta-analysis." Recent patents on inflammation & allergy drug discovery 6, no. 1 (2012): 78-87.

Fritz, Heidi, Deborah A. Kennedy, Mami Ishii, Dean Fergusson, Rochelle Fernandes, Kieran Cooley, and Dugald Seely. Polysaccharide K and Coriolus versicolor extracts for lung cancer: a systematic review. Integrative cancer therapies 14, no. 3 (2015): 201-211.

 

Habtemariam, Solomon. "Trametes versicolor (Synn. Coriolus versicolor) polysaccharides in cancer therapy: Targets and efficacy." Biomedicines 8, no. 5 (2020): 135.

Hobbs C. Medicinal value of turkey tail fungus Trametes versicolor (L.:Fr.) Pilat (aphyllophoromycetideae). Int J Med Mushrooms. 2004; 6:195–218.

Ho, Cheong-Yip, Chi-Fai Kim, Kwok-Nam Leung, Fung Kwok-Pui, Tak-Fu Tse, Helen Chan, and Clara Bik-San Lau. "Differential anti-tumor activity of Coriolus versicolor (Yunzhi) extract through p53-and/or Bcl-2-dependent apoptotic pathway in human breast cancer cells." Cancer biology & therapy 4, no. 6 (2005): 638-644.

Jędrzejewski, Tomasz, Justyna Sobocińska, Małgorzata Pawlikowska, Artur Dzialuk, and Sylwia Wrotek. "Extract from the coriolus versicolor fungus as an anti-inflammatory agent with cytotoxic properties against endothelial cells and breast cancer cells." International Journal of Molecular Sciences 21, no. 23 (2020): 9063.
 

Lau, C. B. S., C. Y. Ho, C. F. Kim, K. N. Leung, K. P. Fung, T. F. Tse, H. H. L. Chan, and M. S. S. Chow. "Cytotoxic activities of Coriolus versicolor (Yunzhi) extract on human leukemia and lymphoma cells by induction of apoptosis." Life Sciences 75, no. 7 (2004): 797-808. 

 

Lu H, Yang Y, Gad E, et al. TLR2 agonist PSK activates human NK cells and enhances the antitumor effect of HER2-targeted monoclonal antibody therapy. Clin Cancer Res. 2011;17(21):6742–6753.

 

Lu H, Yang Y, Gad E, et al. Polysaccharide krestin is a novel TLR2 agonist that mediates inhibition of tumor growth via stimulation of CD8 T cells and NK cells. Clin Cancer Res. 2011;17(1):67–76.

 

Ng, T. B. A review of research on the protein-bound polysaccharide (polysaccharopeptide, PSP) from the mushroom Coriolus versicolor (Basidiomycetes: Polyporaceae). General Pharmacology: The Vascular System 30, no. 1 (1998): 1-4.

 

Ohwada S, et al. Adjuvant immunochemotherapy with oral Tegafur/Uracil plus PSK in patients with stage II or III colorectal cancer: a randomised controlled study. Br J Cancer2004; 90(5): 1003-10.

Pilkington, Karen, L. Susan Wieland, Lida Teng, Xin Yan Jin, Dawn Storey, and Jian Ping Liu. "Coriolus (Trametes) versicolor mushroom to reduce adverse effects from chemotherapy or radiotherapy in people with colorectal cancer." Cochrane Database of Systematic Reviews 11 (2022).

 

Rotolo, G. The effectiveness of Coriolus versicolor in the treatment of secondary phenomena associated with HIV. In 10th International Congress of Mucosal Immunology. 1999.

Saleh, Mohammad H., Iran Rashedi, and Armand Keating. "Immunomodulatory properties of Coriolus versicolor: the role of polysaccharopeptide." Frontiers in immunology 8 (2017): 1087.

 

Serrano, Luis, Andrés Carlos López, Silvia P. González, Santiago Palacios, Damián Dexeus, Cristina Centeno-Mediavilla, Pluvio Coronado et al. "Efficacy of a Coriolus versicolor–based vaginal gel in women with human papillomavirus–dependent cervical lesions: the PALOMA study." Journal of lower genital tract disease 25, no. 2 (2021): 130.

 

Toi M, Hattori T, Akagi M, et al. Randomized adjuvant trial to evaluate the addition of Tamoxifen and Krestin (PSK)™ to chemotherapy in patients with primary breast cancer. 5-Year results from the Nishi-Nippon Group of the Adjuvant Chemoendocrine Therapy for Breast Cancer Organization. Cancer. 1992; 70:2475–2483.

 

Tsang, Kenneth W., C. L. Lam, C. Yan, J. C. Mak, G. C. Ooi, J. C. Ho, B. Lam, R. Man, J. S. Sham, and W. K. Lam. Coriolus versicolor polysaccharide peptide slows progression of advanced non-small cell lung cancer. Respiratory medicine 97, no. 6 (2003): 618-624.

 

Wan JMF, Sit WH, Yang X, Jiang P, Wong LY. Polysaccharopeptides derived from Coriolus versicolor potentiate the S-phase specific cytotoxicity of Camptothecin (CPT) on human leukemia HL-60 cells. Chinese Medicine. 2010; 5:16.

Yang, Cindy Lai-Hung, Stanley Chi-Chung Chik, Allan Sik-Yin Lau, and Godfrey Chi-Fung Chan. "Coriolus versicolor and its bioactive molecule are potential immunomodulators against cancer cell metastasis via inactivation of MAPK pathway." Journal of Ethnopharmacology 301 (2023): 115790.

 

Yang QY. Yun Zhi polysaccharopeptide (PSP) and the general aspects of its research. Fung Sci. 1997; 12:1–8.

 

Yoshikawa R, Yanagi H, Hashimoto-Tamaoki T, Morinaga T, Nakano Y, Noda M, et al. Gene expression in response to anti-tumour intervention by polysaccharide-K (PSK) in colorectal carcinoma cells. Oncol Rep 2004 Dec;12(6):1287-93. 

 

 

Ganoderma lucidum Reishi, Lingzi and other Ganoderma Species

 

Cao QZ and Lin ZB. Antitumor and anti-angiogenic activity of Ganoderma lucidum polysaccharides peptide. Acta Pharmacol Sin 2004; 25: 833-8.

 

Cao, Qi-zhen, and Zhi-Bin Lin. Ganoderma lucidum polysaccharides peptide inhibits the growth of vascular endothelial cell and the induction of VEGF in human lung cancer cell. Life Sciences 78, no. 13 (2006): 1457-1463.

 

Chang YH, Yang JS, Yang JL, et al. Ganoderma lucidum extracts inhibited leukemia WEHI-3 cells in BALB/c mice and promoted an immune response in vivo. Biosci Biotechnol Biochem. 2009;73(12):2589–2594. 

 

Chen, Nian-Hong, Jian-Wen Liu, and Jian-Jiang Zhong. Ganoderic acid T inhibits tumor invasion in vitro and in vivo through inhibition of MMP expression. Pharmacological Reports 62, no. 1 (2010): 150-163.

 

El-Mekkawy, Sahar, Meselhy R. Meselhy, Norio Nakamura, Yasuhiro Tezuka, Masao Hattori, Nobuko Kakiuchi, Kunitada Shimotohno, Takuya Kawahata, and Toru Otake. Anti-HIV-1 and anti-HIV-1-protease substances from Ganoderma lucidum. Phytochemistry 49, no. 6 (1998): 1651-1657.

 

Gao, Yihuai, Wenbo Tang, Xihu Dai, He Gao, Guoliang Chen, Jinxian Ye, Eli Chan, Hwee Ling Koh, Xiaotian Li, and Shufeng Zhou. Effects of water-soluble Ganoderma lucidum polysaccharides on the immune functions of patients with advanced lung cancer. Journal of medicinal food 8, no. 2 (2005): 159-168.

 

Gao, J.J., B.S. Min, E.M. Ahn, N. Nakamura, H.K. Lee and M. Hattori, 2002. New triterpenes aldehydes, lucialadehydes A-C, from Ganoderma lucidum and their cytotoxicity against murine and human tumor cell lines. Chemical and Pharmacological Bulletin Jun; 50(6): 837-40.

 

Gao, Yihuai, Shufeng Zhou, Jianbo Wen, Min Huang, and Anlong Xu. Mechanism of the antiulcerogenic effect of Ganoderma lucidum polysaccharides on indomethacin-induced lesions in the rat. Life sciences 72, no. 6 (2002): 731-745.

Hsu M. J., Lee S. S., Lin W. W. (2002). Polysaccharide purified from Ganoderma luciduminhibits spontaneous and Fas-mediated apoptosis in human neutrophils through activation of the phosphatidylinositol 3 kinase/Akt pathways. J. Leuokoc. Biol. 72, 207–216.

Hsu, Ming‐Jen, Shiuh‐Sheng Lee, Sho Tone Lee, and Wan‐Wan Lin. Signaling mechanisms of enhanced neutrophil phagocytosis and chemotaxis by the polysaccharide purified from Ganoderma lucidum. British journal of pharmacology 139, no. 2 (2003): 289-298.

 

Jiang, Jiahua, Veronika Slivova, Tatiana Valachovicova, Kevin Harvey, and Daniel Sliva. Ganoderma lucidum inhibits proliferation and induces apoptosis in human prostate cancer cells PC-3. International journal of oncology 24, no. 5 (2004): 1093-1099.

 

Jiang, Jiahua, Veronika Slivova, Kevin Harvey, Tatiana Valachovicova, and Daniel Sliva. Ganoderma lucidum suppresses growth of breast cancer cells through the inhibition of Akt/NF-κB signaling. Nutrition and cancer 49, no. 2 (2004): 209-216.

 

Jin, Xingzhong, Julieta Ruiz Beguerie, Daniel Man‐yeun Sze, and Godfrey CF Chan. Ganoderma lucidum (Reishi mushroom) for cancer treatment. Cochrane Database of Systematic Reviews 6 (2012).

 

Joseph S, Sabulal B, George V, Antony KR, Janardhanan KK. Antitumor and anti-inflammatory activities of polysaccharides isolated from Ganoderma lucidum. Acta Pharm. 2011;61(3):335–342.

 

Juan Lu, Jia-Zhang Qin, Ping Chen, Xi Chen, Ying-Zhi Zhang, and Si-Jia Zhao. Quality Difference Study of Six Varieties of Ganoderma lucidum with Different Origins. Frontiers in Pharmacology. 2012; 3: 57.

 

Kohda, Hiroshi, Wakako Tokumoto, Kiyoe Sakamoto, MICHIKO FUJII, Yuko Hirai, Kazuo Yamasaki, Yasuo Komoda, Hideo Nakamura, SHIGEMASA ISHIHARA, and Masaru Uchida. The biologically active constituents of Ganoderma lucidum (Fr.) Karst. Histamine release-inhibitory triterpenes. Chemical and Pharmaceutical Bulletin 33, no. 4 (1985): 1367-1374.

 

Kuo, Mei-Chun; Weng, Ching-Yi; Ha, Choi-Lan; Wu, Ming-Jiuan (2006). Ganoderma lucidum mycelia enhance innate immunity by activating NF-κB. Journal of Ethnopharmacology 103 (2): 217–22. 

 

Lin, Shwu-Bin, Chyi-Hann Li, Shiuh-Sheng Lee, and Lou-Sing Kan. Triterpene-enriched extracts from Ganoderma lucidum inhibit growth of hepatoma cells via suppressing protein kinase C, activating mitogen-activated protein kinases and G2-phase cell cycle arrest. Life sciences 72, no. 21 (2003): 2381-2390.

 

Lin, Zhi-Bin, and Hui-Na Zhang. Anti-tumor and immunoregulatory activities of Ganoderma lucidum and its possible mechanisms. Acta Pharmacologica Sinica 25 (2004): 1387-1395.

 

Lin, Zhi-Bin. Cellular and molecular mechanisms of immuno-modulation by Ganoderma lucidum. Journal of pharmacological sciences 99, no. 2 (2005): 144-153.

 

Liu, Xin, Jian-Ping Yuan, Chee-Keung Chung, and Xiao-Jun Chen. Antitumor activity of the sporoderm-broken germinating spores of Ganoderma lucidum. Cancer letters 182, no. 2 (2002): 155-161.

 

Lu, Qing-Yi, Yu-Sheng Jin, Qifeng Zhang, Zuofeng Zhang, David Heber, Vay Liang W. Go, Frederick P. Li, and Jian Yu Rao. Ganoderma lucidum extracts inhibit growth and induce actin polymerization in bladder cancer cells in vitro. Cancer letters 216, no. 1 (2004): 9-20.

 

Mahajna, Jamal, Nesly Dotan, Ben-Zion Zaidman, Roumyana D. Petrova, and Solomon P. Wasser. Pharmacological values of medicinal mushrooms for prostate cancer therapy: the case of Ganoderma lucidum. Nutrition and cancer 61, no. 1 (2008): 16-26.

 

Martínez-Montemayor MM, Acevedo RR, Otero-Franqui E, Cubano LA, Dharmawardhane SF. Ganoderma lucidum (Reishi) inhibits cancer cell growth and expression of key molecules in inflammatory breast cancer. Nutr Cancer. 2011;63(7):1085–1094.

 

Min, Byung-Sun, Jiang-Jing GAo, Norio Nakamura, and Masao Hattori. Triterpenes from the spores of Ganoderma lucidum and their cytotoxicity against meth-A and LLC tumor cells. Chemical and Pharmaceutical Bulletin 48, no. 7 (2000): 1026-1033.

 

Miyazaki, Toshio, and Motohiro Nishijima. Studies on fungal polysaccharides. XXVII. Structural examination of a water-soluble, antitumor polysaccharide of Ganoderma lucidum. Chemical and Pharmaceutical Bulletin 29, no. 12 (1981): 3611-3616.

 

Morigiwa, Aiko, Katsuaki Kitabatake, Yoshinori Fujimoto, and Nobuo Ikekawa. Angiotensin converting enzyme-inhibitory triterpenes from Ganoderma lucidim. Chemical and Pharmaceutical Bulletin 34, no. 7 (1986): 3025-3028.

 

Müller, Claudia I., Takashi Kumagai, James O’Kelly, Navindra P. Seeram, David Heber, and H. Phillip Koeffler. Ganoderma lucidum causes apoptosis in leukemia, lymphoma and multiple myeloma cells. Leukemia research 30, no. 7 (2006): 841-848.

 

Paterson, R. Russell M. Ganoderma–a therapeutic fungal biofactory. Phytochemistry 67, no. 18 (2006): 1985-2001.

 

Sanodiya, Bhagwan S., Gulab S. Thakur, Rakesh K. Baghel, G. B. K. S. Prasad, and P. S. Bisen. Ganoderma lucidum: a potent pharmacological macrofungus. Current pharmaceutical biotechnology 10, no. 8 (2009): 717-742.

Sliva, Daniel (2003). Ganoderma Lucidum (Reishi) in Cancer Treatment. Integrative Cancer Therapies 2 (4): 358–64.

 

Sliva, Daniel, Carlos Labarrere, Veronika Slivova, Miroslav Sedlak, Frank P. Lloyd Jr, and Nancy WY Ho. Ganoderma lucidum suppresses motility of highly invasive breast and prostate cancer cells. Biochemical and biophysical research communications 298, no. 4 (2002): 603-612.

 

Sone, Yoshiaki, Reiko Okuda, Noriko Wada, Etsu Kishida, and Akira Misaki. Structures and antitumor activities of the polysaccharides isolated from fruiting body and the growing culture of mycelium of Ganoderma lucidum. Agricultural and biological chemistry 49, no. 9 (1985): 2641-2653.

 

Stanley, Gwenaelle, Kevin Harvey, Veronika Slivova, Jiahua Jiang, and Daniel Sliva. Ganoderma lucidum suppresses angiogenesis through the inhibition of secretion of VEGF and TGF-β1 from prostate cancer cells. Biochemical and biophysical research communications 330, no. 1 (2005): 46-52.

 

Suarez-Arroyo IJ, Rosario-Acevedo R, Aguilar-Perez A, Clemente PL, Cubano LA, Serrano J, et al. (2013) Anti-Tumor Effects of Ganoderma lucidum (Reishi) in Inflammatory Breast Cancer in In Vivo and In Vitro Models. PLoS ONE 8(2): e57431. doi:10.1371/journal.pone.0057431.

Tang, Wen, Jian-Wen Liu, Wei-Ming Zhao, Dong-Zhi Wei, and Jian-Jiang Zhong. Ganoderic acid T from Ganoderma lucidum mycelia induces mitochondria mediated apoptosis in lung cancer cells. Life sciences 80, no. 3 (2006): 205-211.

 

Unlu, Ahmet, Erdinc Nayir, Onder Kirca, and Mustafa Ozdogan. Ganoderma lucidum (reishi mushroom) and cancer. J. BUON 21 (2016): 792-798.

 

Wachtel-Galor, Sissi, John Yuen, John A. Buswell, and Iris FF Benzie. Ganoderma lucidum (Lingzhi or Reishi). In Herbal Medicine: Biomolecular and Clinical Aspects. 2nd edition. CRC Press/Taylor & Francis, 2011.

 

Wang PY, Zhu XL, Lin ZB. Antitumor and immunomodulatory effects of polysaccharides from broken-spore of Ganoderma lucidum. Front Pharmacol. 2012 Jul;3:135.

 

Wang, Sheng‐Yuan, Ming‐Ling Hsu, Hui‐Chi Hsu, Shiuh‐Sheng Lee, Ming‐Shi Shiao, and Chi‐Kuan Ho. The anti‐tumor effect of Ganoderma lucidum is mediated by cytokines released from activated macrophages and T lymphocytes. International journal of cancer 70, no. 6 (1997): 699-705.

 

Wang G, Zhao J, Liu J, Huang Y, Zhong JJ, Tang W. Enhancement of IL-2 and IFN-gamma expression and NK cells activity involved in the anti-tumor effect of ganoderic acid Me in vivo. Int Immunopharmacol. 2007;7(6):864–870.

 

Weng, Chia-Jui, and Gow-Chin Yen. The in vitro and in vivo experimental evidences disclose the chemopreventive effects of Ganoderma lucidum on cancer invasion and metastasis. Clinical & experimental metastasis 27, no. 5 (2010): 361-369.

 

Xie, J. T., C. Z. Wang, S. Wicks, J. J. Yin, J. Kong, J. Li, Y. C. Li, and C. S. Yuan. Ganoderma lucidum extract inhibits proliferation of SW 480 human colorectal cancer cells. Experimental Oncology (2006).

Yuen, John; Gohel, Mayur Danny (2005). Anticancer Effects of Ganoderma lucidum: A Review of Scientific Evidence. Nutrition and Cancer 53 (1): 11–7. 

Zhang, Hui-Na, and Zhi-Bin Lin. Hypoglycemic effect of Ganoderma lucidum polysaccharides. Acta Pharmacologica Sinica 25, no. 2 (2004): 191-195.

 

Zhu XL, Chen AF, Lin ZB. Ganoderma lucidum polysaccharides enhance the function of immunological effector cells in immunosuppressed mice. J Ethnopharmacol. 2007;111(2):219–226.

 

Grifola frondosa Maitake

 

Alexander, Bobby, Andrew I. Fishman, Majid Eshghi, Muhammad Choudhury, and Sensuke Konno. Induction of cell death in renal cell carcinoma with combination of D-fraction and vitamin C. Integrative cancer therapies 12, no. 5 (2013): 442-448.

 

Alonso, Eliana Noelia, Manuela Orozco, Alvaro Eloy Nieto, and Gabriela Andrea Balogh. Genes related to suppression of malignant phenotype induced by Maitake D-Fraction in breast cancer cells. Journal of medicinal food 16, no. 7 (2013): 602-617.

 

Deng G, Lin H, Seidman A, et al. (September 2009). A phase I/II trial of a polysaccharide extract from Grifola frondosa (Maitake mushroom) in breast cancer patients: immunological effects. Journal of Cancer Research and Clinical Oncology 135 (9): 1215–21.

 

Harada N, Kodama N, Nanba H. Relationship between dendritic cells and the D-fraction-induced Th-1 dominant response in BALB/c tumor-bearing mice. Cancer Lett. 2003;192(2):181–187.

 

Horio H, Ohtsuru M. Maitake (Grifola frondosa) improve glucose tolerance of experimental diabetic rats. J Nutr Sci Vitaminol 2001; 47:57-63.

 

Inoue A, Kodama N, Nanba H. Effect of maitake (Grifola frondosa) D-fraction on the control of the T lymph node Th-1/Th-2 proportion. Biol Pharm Bull. 2002;25(4):536–540. 

 

Kodama N, Komuta K, Sakai N, Nanba H (December 2002). Effects of D-Fraction, a polysaccharide from Grifola frondosa on tumor growth involves activation of NK cells. Biological & Pharmaceutical Bulletin 25 (12): 1647–50.   

 

Kodama, Noriko, Maho Yamada, and Hiroaki Nanba. Addition of Maitake D-fraction reduces the effective dosage of vancomycin for the treatment of Listeria-infected mice. The Japanese Journal of Pharmacology 87, no. 4 (2001): 327-332.

Kubo K, Aoki H. Nanba H. Anti-diabetic activity present in the fruit body of Grifola frondosa (Maitake). Biol Pharm Bull 1994; 17:1106-10.

 

Lin H, de Stanchina E, Zhou XK, et al. Maitake beta-glucan promotes recovery of leukocytes and myeloid cell function in peripheral blood from paclitaxel hematotoxicity. Cancer Immunol Immunother. 2010 Jun;59(6):885-97.

 

Lin, Hong, Sandy WY Cheung, Mirjana Nesin, Barrie R. Cassileth, and Susanna Cunningham-Rundles. Enhancement of umbilical cord blood cell hematopoiesis by maitake beta-glucan is mediated by granulocyte colony-stimulating factor production. Clin. Vaccine Immunol. 14, no. 1 (2007): 21-27.

Louie, Brandon, Srinivas Rajamahanty, John Won, Muhammad Choudhury, and Sensuke Konno. Synergistic potentiation of interferon activity with maitake mushroom d‐fraction on bladder cancer cells. BJU international 105, no. 7 (2010): 1011-1015.

Masuda Y, Inoue H, Ohta H, Miyake A, Konishi M, Nanba H. Oral administration of soluble β-glucans extracted from Grifola frondosa induces systemic antitumor immune response and decreases immunosuppression in tumor-bearing mice. Int J Cancer. 2013;133(1):108–119

 

Masuda Y, Murata Y, Hayashi M, Nanba H. Inhibitory effect of MD-Fraction on tumor metastasis: involvement of NK cell activation and suppression of intercellular adhesion molecule (ICAM)-1 expression in lung vascular endothelial cells. Biol Pharm Bull 2008 Jun;31(6):1104-8.

 

Pyo P, Louie B, Rajamahanty S, Choudhury M, Konno S. Possible immunotherapeutic potentiation with D-fraction in prostate cancer cells. J Hematol Oncol. 2008 Dec;1:25. 

 

Soares, Raquel, Manuela Meireles, Ana Rocha, Ana Pirraco, Diego Obiol, Eliana Alonso, Gisela Joos, and Gabriela Balogh. Maitake (D fraction) mushroom extract induces apoptosis in breast cancer cells by BAK-1 gene activation. Journal of medicinal food 14, no. 6 (2011): 563-572.
 

Wesa, Kathleen M., Susanna Cunningham-Rundles, Virginia M. Klimek, Emily Vertosick, Marci I. Coleton, K. Simon Yeung, Hong Lin, Stephen Nimer, and Barrie R. Cassileth. Maitake mushroom extract in myelodysplastic syndromes (MDS): a phase II study. Cancer Immunology, Immunotherapy 64, no. 2 (2015): 237-247.

 

Hericium erinaceus Yamabushitake

 

Abdulla, Mahmood Ameen, Suzita Noor, Kah-Hui Wong, and Hapipah Mohd Ali. Effect of culinary-medicinal lion's mane mushroom, Hericium erinaceus (Bull.: Fr.) Pers.(Aphyllophoromycetideae), on ethanol-induced gastric ulcers in rats. International Journal of Medicinal Mushrooms 10, no. 4 (2008).

 

Chaiyasut, Chaiyavat, and Bhagavathi Sundaram Sivamaruthi. Anti-hyperglycemic property of Hericium erinaceus–A mini review. Asian Pacific Journal of Tropical Biomedicine 7, no. 11 (2017): 1036-1040.

 

Friedman, Mendel. Chemistry, nutrition, and health-promoting properties of Hericium erinaceus (lion’s mane) mushroom fruiting bodies and mycelia and their bioactive compounds. Journal of agricultural and food chemistry 63, no. 32 (2015): 7108-7123.

 

Hazekawa, Mai, Aiko Kataoka, Kazuhide Hayakawa, Takeshi Uchimasu, Riyo Furuta, Keiichi Irie, Yoshiharu Akitake et al. Neuroprotective effect of repeated treatment with Hericium erinaceum in mice subjected to middle cerebral artery occlusion. Journal of Health Science 56, no. 3 (2010): 296-303.

 

He, Xirui, Xiaoxiao Wang, Jiacheng Fang, Yu Chang, Ning Ning, Hao Guo, Linhong Huang, Xiaoqiang Huang, and Zefeng Zhao. Structures, biological activities, and industrial applications of the polysaccharides from Hericium erinaceus (Lion’s Mane) mushroom: A review. International journal of biological macromolecules 97 (2017): 228-237.

 

Hiwatashi, Kazuyuki, Yasuyuki Kosaka, Nao Suzuki, Keishi Hata, Toshiyuki Mukaiyama, Kenji Sakamoto, Hitoshi Shirakawa, and Michio Komai. Yamabushitake mushroom (Hericium erinaceus) improved lipid metabolism in mice fed a high-fat diet. Bioscience, biotechnology, and biochemistry 74, no. 7 (2010): 1447-1451.

 

Kolotushkina, E. V., M. G. Moldavan, K. Yu Voronin, and G. G. Skibo. The influence of Hericium erinaceus extract on myelination process in vitro. Fiziol Zh 49, no. 1 (2003): 38-45.

 

Mori, Koichiro, Yutaro Obara, Takahiro Moriya, Satoshi Inatomi, and Norimichi Nakahata. Effects of Hericium erinaceus on amyloid β (25-35) peptide-induced learning and memory deficits in mice. Biomedical Research 32, no. 1 (2011): 67-72.

Mori, Koichiro, Yutaro Obara, Mitsuru Hirota, Yoshihito Azumi, Satomi Kinugasa, Satoshi Inatomi, and Norimichi Nakahata. Nerve growth factor-inducing activity of Hericium erinaceus in 1321N1 human astrocytoma cells. Biological and Pharmaceutical Bulletin 31, no. 9 (2008): 1727-1732.

 

Nagano, Mayumi, Kuniyoshi Shimizu, Ryuichiro Kondo, Chickako Hayashi, Daigo Sato, Katsuyuki Kitagawa, and Koichiro Ohnuki. Reduction of depression and anxiety by 4 weeks Hericium erinaceus intake. Biomedical Research 31, no. 4 (2010): 231-237.

 

Phan, Chia-Wei, Guan-Serm Lee, Sok-Lai Hong, Yuin-Teng Wong, Robert Brkljača, Sylvia Urban, Sri Nurestri Abd Malek, and Vikineswary Sabaratnam. Hericium erinaceus (Bull.: Fr) Pers. cultivated under tropical conditions: isolation of hericenones and demonstration of NGF-mediated neurite outgrowth in PC12 cells via MEK/ERK and PI3K-Akt signaling pathways. Food & function 5, no. 12 (2014): 3160-3169.

 

Phan, Chia-Wei, Pamela David, Murali Naidu, Kah-Hui Wong, and Vikineswary Sabaratnam. Therapeutic potential of culinary-medicinal mushrooms for the management of neurodegenerative diseases: diversity, metabolite, and mechanism. Critical Reviews in Biotechnology 35, no. 3 (2015): 355-368.

 

Samberkar, Snehlata, Sivasangkary Gandhi, Murali Naidu, Kah-Hui Wong, Jegadeesh Raman, and Vikineswary Sabaratnam. Lion's Mane, Hericium erinaceus and Tiger Milk, Lignosus rhinocerotis (Higher Basidiomycetes) medicinal mushrooms stimulate neurite outgrowth in dissociated cells of brain, spinal cord, and retina: an in vitro study. International journal of medicinal mushrooms 17, no. 11 (2015).

 

Thongbai, Benjarong, Sylvie Rapior, Kevin D. Hyde, Kathrin Wittstein, and Marc Stadler. Hericium erinaceus, an amazing medicinal mushroom. Mycological Progress 14, no. 10 (2015): 91.

 

Wong, Kah-Hui, Murali Naidu, Rosie Pamela David, Robiah Bakar, and Vikineswary Sabaratnam. Neuroregenerative potential of lion's mane mushroom, Hericium erinaceus (Bull.: Fr.) Pers.(higher Basidiomycetes), in the treatment of peripheral nerve injury. International journal of medicinal mushrooms 14, no. 5 (2012).

 

Wong, Jing-Yang, Mahmood Ameen Abdulla, Jegadeesh Raman, Chia-Wei Phan, Umah Rani Kuppusamy, Shahram Golbabapour, and Vikineswary Sabaratnam. Gastroprotective effects of Lion’s Mane mushroom Hericium erinaceus (Bull.: Fr.) Pers.(Aphyllophoromycetideae) extract against ethanol-induced ulcer in rats. Evidence-Based Complementary and Alternative Medicine 2013 (2013).

 

Inonotus obliquus Chaga

 

Chung, Mi Ja, Cha-Kwon Chung, Yoonhwa Jeong, and Seung-Shi Ham. Anticancer activity of subfractions containing pure compounds of Chaga mushroom (Inonotus obliquus) extract in human cancer cells and in Balbc/c mice bearing Sarcoma-180 cells. Nutrition research and practice 4, no. 3 (2010): 177-182.

 

Cui Y, Kim DS, Park KC. Antioxidant effect of Inonotus obliquus. J Ethnopharmacol. 2005 Jan 4;96(1-2):79-85.

Ham SS, Kim SH, Moon SY, Chung MJ, Cui CB, Han EK, Chung CK, Choe M. Antimutagenic effects of subfractions of Chaga mushroom (Inonotus obliquus) extract. Mutat Res 2009; 672:55–59.

 

Kim, Min-A., Yong-Seob Jeong, Gei-Taek Chun, and Youn-Soo Cha. Antihyperlipidemic and glycemic control effects of mycelia of inonotus obliquus including protein-bound polysaccharides extract in C57BL/6J mice. Journal of the Korean Society of Food Science and Nutrition 38, no. 6 (2009): 667-673.

 

Lee SH, Hwang HS, Yun JW. Antitumor activity of water extract of a mushroom, Inonotus obliquus, against HT-29 human colon cancer cells. Phytother Res. Apr 15 2009.

 

Mazurkiewicz, W. I. T. O. L. D., Katarzyna Rydel, D. A. R. I. U. S. Z. Pogocki, Marta Kinga Lemieszek, Ewa Langner, and W. O. J. C. I. E. C. H. Rzeski. Separation of an aqueous extract Inonotus obliquus (Chaga). A novel look at the efficiency of its influence on proliferation of A549 human lung carcinoma cells. Acta Pol Pharm 67, no. 4 (2010): 397-406.

 

Nomura, Masaaki, Tatsuo Takahashi, Aimi Uesugi, Reiko Tanaka, and Shinjiro Kobayashi. Inotodiol, a lanostane triterpenoid, from Inonotus obliquus inhibits cell proliferation through caspase-3-dependent apoptosis. Anticancer research 28, no. 5A (2008): 2691-2696.

 

Shibnev, V. A., D. V. Mishin, T. M. Garaev, N. P. Finogenova, A. G. Botikov, and P. G. Deryabin. Antiviral activity of Inonotus obliquus fungus extract towards infection caused by hepatitis C virus in cell cultures. Bulletin of experimental biology and medicine 151, no. 5 (2011): 612-614.

 

Song, Fu-Qiang, Ying Liu, Xiang-Shi Kong, Wei Chang, and Ge Song. Progress on understanding the anticancer mechanisms of medicinal mushroom: Inonotus obliquus. Asian Pacific Journal of Cancer Prevention 14, no. 3 (2013): 1571-1578.

 

Won, Dong Pil, Jong Seok Lee, Duck Soo Kwon, Keun Eok Lee, Won Cheol Shin, and Eock Kee Hong. Immunostimulating activity by polysaccharides isolated from fruiting body of Inonotus obliquus. Molecules and cells 31, no. 2 (2011): 165-173.

 

Yang, Byung-keun, Kai-Yip Cho, Michael A. Wilson, and Chi-Hyun Song. Effects of Inonotus obliquus mycelia on the level of plasma glucose and lipids in streptozotocin-induced diabetic rats. The Korean journal of mycology 33, no. 2 (2005): 64-68.

 

Youn MJ, Kim JK, Park SY, et al. Chaga mushroom (Inonotus obliquus) induces G0/G1 arrest and apoptosis in human hepatoma HepG2 cells. World J Gastroenterol. Jan 28 2008;14(4):511-517.

 

 

Lentinus edodes Shiitake 

 

Bisen, P. S., Rakesh K. Baghel, Bhagwan S. Sanodiya, Gulab S. Thakur, and G. B. K. S. Prasad. Lentinus edodes: a macrofungus with pharmacological activities. Current Medicinal Chemistry 17, no. 22 (2010): 2419-2430.

 

Finimundy, Tiane Cristine, Aldo José Pinheiro Dillon, João Antônio Pêgas Henriques, and Mariana Roesch Ely. A review on general nutritional compounds and pharmacological properties of the Lentinula edodes mushroom. Food and Nutrition Sciences 5, no. 12 (2014): 1095.

 

Hobbs, Christopher. Medicinal value of Lentinus edodes (Berk.) Sing.(Agaricomycetideae). A literature review. International Journal of Medicinal Mushrooms 2, no. 4 (2000). 

 

Ng, Mah-Lee, and Ann-Teck Yap. Inhibition of human colon carcinoma development by lentinan from shiitake mushrooms (Lentinus edodes). The Journal of Alternative & Complementary Medicine 8, no. 5 (2002): 581-589.

 

Oh, Se-In, and Mee-Sook Lee. Antioxidative stress and antimutagenic effects of Lentinus edodes ethanol extracts. The Korean Journal of Food And Nutrition 20, no. 4 (2007): 341-348.

Click for links

Agaricus Blazei Murill
cordyceps harvesting
Ganoderma lucidum
Coriolus versicolor
inonotus obliquus (chaga)
bottom of page