Skip to main content

Current evidence for the hepatoprotective activities of the medicinal mushroom Antrodia cinnamomea

Abstract

Antrodia cinnamomea (AC) is an endemic mushroom species of Taiwan, and has been demonstrated to possess diverse biological and pharmacological activities, such as anti-hypertension, anti-hyperlipidemia, anti-inflammation, anti-oxidation, anti-tumor, and immunomodulation. This review focuses on the inhibitory effects of AC on hepatitis, hepatocarcinoma, and alcohol-induced liver diseases (e.g., fatty liver, fibrosis). The relevant biochemical and molecular mechanisms are addressed. Overall, this review summarizes the hepatoprotective activities in vitro and in vivo. However, there is no doubt that human and clinical trials are still limited, and further studies are required for the development of AC-related products.

Introduction

Antrodia cinnamomea (AC) (niu-chang-chih in Chinese) is a medicinal mushroom (Figure 1) endemic to Taiwan. It has been well-known for its medicinal properties, particularly with regard to liver complaints [1], since its first identification as a new species by Zang and Su in 1990 [2]. Among its diverse pharmacological activities, the evidence for hepatic protection is the most recognized and the strongest. Studies have shown that AC can inhibit hepatic tumor growth and retard the progression of hepatitis [3, 4]. Although AC is currently used as a food supplement, the Food and Drug Administration (FDA) has not approved any AC extracts or purified compounds for clinical applications. Some AC products are claimed to protect the liver against food and drug intoxication, especially alcohol-induced liver damage, maintain hepatic homeostasis, or both. This article reviews the current evidence for the hepatoprotective properties (such as effects on hepatitis, cirrhosis, liver cancers, and alcoholic damage) of AC extracts and active compounds.

Figure 1
figure 1

Morphological appearance of Antrodia cinnamomea .

Effects on hepatitis

General background of hepatitis

Hepatitis is one of the top ten causes of death worldwide (more than one million deaths per year), and is mainly classified into hepatitis A, B, and C, involving different types of viruses, epigenetic and genetic alterations, pathological progressions, and clinical treatments [5]. Hepatitis A is an acute infectious liver disease caused by hepatitis A virus (HAV). It is transmitted by a fecal-to-oral route, and mainly occurs as outbreaks from fecal contamination of food or water sources or direct contact with carriers. It has no clinical signs or symptoms in most infected people, and most cases recover within 1–2 months [6]. Hepatitis A causes the least problems among the various types of hepatitis, and its incidence rate has been diminishing in recent years, probably through improved hygiene [6]. Hepatitis B and C are acute and chronic infections caused by hepatitis B virus (HBV) and hepatitis C virus (HCV), respectively, and are more serious. HBV and HCV belong to the Hepadnaviridae and Flaviviridae, respectively. HBV is a DNA virus that can integrate into the host genome after infection, while HCV is an RNA virus that has no such ability [6]. According to a worldwide epidemiological evaluation, at least 400 and 170 million people are chronically infected with HBV and HCV, respectively [7, 8]. In fact, these viruses persist in the liver in about 80% of infected people [9], and such chronic viral infections may be associated with chronic inflammation that leads to scarring of the liver tissues and cirrhosis [10]. Upon infection, HBV and HCV interact with host hepatocytes, impair cell metabolism (e.g., induce cell death) [11], modulate signaling pathways of protein synthesis (e.g., produce viral proteins and enzymes) [12, 13] and immunity (e.g., synthesize and place viral antigens on host cell surfaces) [14], and develop immortalization (e.g., modulate surface markers). Ultimately, 1–5% of cirrhotic livers develop hepatocellular carcinoma (HCC) [15].

Anti-hepatitis activity

Inhibition of viral proteases, which are crucial for viral replication, can interrupt viral protein maturation. Antrodins (A to E) and their metabolic analogues selectively inhibited HCV NS3/4A protease activity [3]. Among these, antrodin A (the major in vivo metabolite of antrodin C) showed the highest potency with an IC50 of 0.9 μg/mL, while antrodin B showed lower potency with an IC50 of > 100 μg/mL. Moreover, polysaccharides isolated from fruiting bodies and cultured mycelia of AC inhibited HBV replication activity [16]. After an HBV-producing cell line (MS-G2) was treated with the AC-extracted polysaccharides, anti-hepatitis B surface antigen (HBsAg) and anti-hepatitis B e antigen (HBeAg) were reduced [16]. Moreover, the AC-extracted polysaccharides (50 μg/mL) were found to be more effective than interferon-α (IFN-α) (1000 U/mL) for HBsAg and HBeAg inhibition [16]. Huang et al. [17] showed that 2,2′,5,5′-tetramethoxy-3,4,3′,4′-bis(methlylene-dioxy)-6,6′-dimethylbiphenyl (50 μM) could suppress the HBsAg and HBeAg levels in a wild-type HBV-producing cell line (ES2) within a non-toxic range and in a dose-dependent manner. These findings indicate that AC can effectively attenuate hepatitis virus-induced damage by inhibiting essential viral enzyme activities and antigen production.

Effects on HCC

Pathogenesis of HCC

Liver cancer comprises diverse hepatic neoplasms, including HCC, cholangiocarcinoma, hepatoblastoma, and hemangiosarcoma [18, 19]. Among these, HCC is the primary malignant hepatic cancer worldwide, the fifth most common cancer, and the third leading cause of cancer death around the world [20–22]. The incidence and mortality of HCC are both increasing [23–26]. HCC arises in the context of chronic viral hepatitis (e.g., HBV, HCV, and HDV infection), cirrhosis, alcoholism, toxin or drug poisoning (e.g., aflatoxins), and metabolic disorders (e.g., diabetes, non-alcoholic fatty liver disease, and hereditary diseases) [27]. Recently, geographical differences in HCC incidence have been related to the changing distribution and natural history of HBV and HCV infections [28]. The World Health Organization (WHO) reported that HBV is a human carcinogen, and that an estimated 300–400 million individuals are chronically infected with HBV worldwide [29]. Moreover, a large number of carriers fail to eliminate the virus, and progress to chronic viral infection. Continuous hepatic inflammation can lead to chronic hepatitis and cirrhosis. The incidence of HCC in chronically HBV-infected or cirrhotic individuals is 100- and 1000-fold higher, respectively, than in uninfected individuals [27, 30]. For toxin-induced HCC, ingestion of aflatoxin-B1 (a fungal toxin classified as a type I carcinogen by the International Agency for Research on Cancer (IARC) and produced by the molds Aspergillus flavus and Aspergillus parasiticus found in contaminated food) increased the risk of HCC development [31, 32]. Aflatoxin-B1 also induced mutations in codon 249 of the p53 tumor suppressor gene and the RAS oncogene [33–36]. Alcohol, which is also classified as a type I carcinogen, is another critical HCC risk factor [31, 32]. Alcohol activates monocytes and induces pro-inflammatory cytokine production, and the subsequent increase in endotoxins activates Kupffer cells to release chemokines and cytokines (e.g., tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6)). The resulting oxidative stress activates stellate cells to synthesize large amounts of collagen, leading to fibrosis, cirrhosis, and ultimately HCC [37–39]. Apart from these etiological agents, inflammation, oxidative stress, genetic alternations, activation of oncogenes, inactivation of tumor suppressor genes, genomic instability, and overexpression of growth and angiogenic factors are related to the pathogenesis of HCC [31, 40].

Angiogenesis plays an important role in the pathogenesis (i.e., tumor growth, invasion, and metastasis) of all types of solid tumors, including HCC [41]. As a hypervascular tumor, HCC develops and progresses from a small, avascular, and well-differentiated HCC to a large, highly vascular, and poorly differentiated HCC within a short period [42]. The expanded endothelium provides a channel for HCC cells to enter the blood circulation system and metastasize to different tissues. During tumor angiogenesis, angiogenic factors (e.g., vascular endothelial cell growth factor (VEGF), fibroblast growth factor-2 (FGF-2), angiogenin, or angiopoietins) and their receptors are produced by tumor cells and endothelial cells (ECs) [43, 44]. Through VEGF signaling, the most common and potent angiogenic signaling pathway, ECs express VEGF receptors (VEGFR-1 and -2), receive signals from tumor-generated VEGF, proliferate, migrate toward tumor tissues, and form functional neovessels [43, 44]. Moreover, these angiogenic factors induce gene transcriptional, biochemical, and enzymatic activities, such as extracellular digestion of extracellular matrix by matrix metalloproteinases (MMPs), cell cycle progression, and production of bioactive molecules (e.g., angiogenic factors, cytokines, and chemokines) [43, 44]. As a result, both tumor cells and ECs are activated in a feedback mechanism, thereby facilitating tumorigenesis [45].

Anti-hepatocarcinoma activity

Various types of AC extracts and purified components (e.g., antcin A, B, and C, antroquinonol, methyl antcinate A (MAA), methyl antcinate B (MAB) ethylacetate, and methanolic, ethanolic and fermented extracts) exhibit cytotoxicity through apoptosis in various cancers, especially hepatic carcinoma. Apoptosis is processed sequentially through the activation of death receptors (e.g., Fas receptor) and downstream caspases (e.g., caspase-3, -8, and -9), and mitochondrial function disruption. MAA (30.2–78 μM) and antcins A and C (30.2–286.4 μM) stimulate a series of apoptotic pathway cascades, including caspase-2, -3, and -9 activation, Bax, Bak, and PUMA protein expression, Bcl-(XL) and Bcl-2 suppression, and cytochrome c release, resulting in apoptosis [46–48]. MAB and antcin B (100 μM)-induced apoptosis is characterized by increased DNA fragmentation, cleavage of PARP, sub-G1 population appearance, and chromatin condensation [49]. Moreover, AC components induced NADPH oxidase-provoked oxidative stress [49, 50]. Another purified AC component, antroquinonol, inhibited HepG2 (IC50: 0.13 μM) and Hep3B (IC50: 4.3 μM) cell growth [50, 51]. Recently, Chiang et al. [52] demonstrated that antroquinonol induced HCC apoptosis through AMPK activation and mTOR translational pathway inhibition, and induced cell cycle arrest through downregulation and suppressed nuclear translocation of cyclin D1, cyclin E, Cdk4, and Cdk2. An AC ethyl acetate extract (IC50: 42.5 μg/mL in HepG2 cells; 78.3 μg/mL in Hep3B cells), methanolic extract (IC50: 49.5 μg/mL in HepG2 cells; 62.7 μg/mL in Hep3B cells), and ethanolic extract (IC50: 54.2 μg/mL in HepG2 cells; 82.9 μg/mL in Hep3B cells) could also inhibit growth in a similar manner [4, 46, 48, 53, 54]. Apart from the direct cytotoxicity, cell cycle arrest can be used for controlling tumor growth. During the G1 phase of the cell cycle, the cells increase in size and synthesize essential proteins for the resting steps of the cell division process. It is important to control whether a cell goes into the S phase (DNA synthesis phase) or escapes from the cell cycle. An AC mycelium extract, 4-acetylantroquinonol B (EC50: 0.1 μg/ mL), induced cell cycle arrest in HepG2 cells, indicating that cells were unable to divide. Western blotting analyses further showed that the protein expressions of different cell cycle-regulating proteins, CDK2 and CDK4, were decreased, while p21, p27, and p53 were increased in HepG2 cells after the AC treatment [55, 56]. Similar to the case for the conventional anti-cancer drug cisplatin, tumor cell proliferation can be inhibited through induction of DNA damage, cell cycle arrest, and apoptosis. This suppresses the size of tumor tissues, and reduces the chance for subsequent metastasis [57, 58].

Anti-angiogenic therapy has been explored as a therapeutic approach to HCC [59, 60]. A series of angiogenic inhibitors, e.g., bevacizumab (for blocking the angiogenic factor VEGF), endostatin, IFN-α, cyclooxygenase-2 (for inhibiting EC migration, tube formation, proliferation, and angiogenic factor production), interleukin-12 (for producing anti-angiogenic factors), and erlotinib and gefitinib (for inhibiting epidermal growth factor receptor tyrosine kinase activation) are currently in different phases of clinical trials [61].

An AC ethyl acetate extract and AC polysaccharides inhibited angiogenesis in vitro and in vivo. The ethyl acetate extract was shown to inhibit cancer invasion of human liver cancer PLC/PRF/5 cells through suppression of angiogenic factor (e.g. VEGF) and extracellular digestive enzyme (e.g. MMP-2 and -9) production, and elevation of enzyme inhibitor (tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2) production in the concentration ranges of 10 to 40 μg/mL [47]. These results were confirmed in an animal model by Matrigel plug angiogenesis assays, in which the ethyl acetate extract (300 mg/kg) significantly suppressed tumor growth in nude mice [46]. The VGEF-induced tube formation and migration of ECs, and neovessel formation on chorioallantoic membranes were effectively inhibited by the AC polysaccharide extract [62, 63]. Cheng et al. [64] demonstrated that the different fractionated AC polysaccharide extracts were able to inhibit VEGF protein expression, binding of VEGF to VEGFR-2, and VEGFR-2 phosphorylation, suggesting that inhibition of VEGF interactions with VEGF receptors is an anti-angiogenic mechanism of AC.

Anti-angiogenic therapy is particularly promising, because it is expected to minimize the devastating side effects, such as hair loss, bleeding, and immunosuppression, that are commonly associated with conventional chemotherapy. We previously found that many herbal medicines and natural compounds, such as ginsenosides, indirubin derivatives, and sinomenine, possess anti-angiogenic activities [65–70]. Therefore, AC may also contain anti-angiogenic components.

Effects on alcohol metabolism

Metabolism of alcohol and its related diseases

Chronic excessive consumption of alcohol often causes serious health problems, especially to the liver, such as cirrhosis and HCC [18, 71]. According to the WHO, alcohol is the third largest risk factor for disease worldwide, and has been classified as a type I carcinogen by the IARC [72]. It has been estimated that alcohol leads to 2.5 million deaths each year, and that 320,000 of these deaths are in the age group of 15–29 years, representing 9% of all deaths in that age group [73]. Alcohol, a water-soluble small molecule, is easily absorbed into the bloodstream from the gastrointestinal tract. It cannot be stored, and is either metabolized or excreted [74, 75]. More than 90% of absorbed alcohol is oxidized by hepatocytes, while the remainder is excreted through pulmonary respiration and urinary excretion [75, 76]. There are three major pathways for metabolizing alcohol. The primary pathway of alcohol metabolism is the alcohol dehydrogenase (ADH)-mediated ethanol oxidation pathway [76, 77]. ADH comprises a group of cytosolic enzymes that convert alcohol to acetaldehyde utilizing NAD+ as a coenzyme, and the elements of this pathway are mainly located in the liver. At high alcohol concentrations (above 100 mg/dL) or during chronic alcohol consumption, the ADH pathway may become saturated through NAD+ depletion. When this happens, the secondary pathway, the microsomal ethanol oxidizing system, is used to break down alcohol [76, 77]. In this system, cytochrome P450 enzymes such as CYP2E1, CYP1A2, and CYP2A4 metabolize alcohol to acetaldehyde via NADPH as a coenzyme instead of NAD+[76, 78]. The third pathway is the hydrogen peroxide (H2O2)-generating system in peroxisomes [76], which oxidizes alcohol to acetaldehyde and water molecules [75, 77]. The acetaldehyde product is further oxidized by aldehyde dehydrogenase (ALDH) to acetate, and subsequently processed in the citric acid cycle. The final products are carbon dioxide, water, and ATP. Through the ADH and ALDH-mediated reactions, NAD+ is used up in the form of a coenzyme, and NADH is generated. This increased ratio of NADH:NAD+ redox potential in the liver inhibits mitochondrial β-oxidation of fatty acids and reduces lipid expenditure, resulting in lipogenesis and lipid accumulation in hepatocytes [79, 80]. The severity of this alcoholic liver disease (ALD) depends on the amount, frequency, and duration of alcohol consumption, and can further develop into more complicated liver damage states, including fibrosis, steatohepatitis, cirrhosis, and cancer [81–83].

Anti-alcoholic activity

Different types of AC preparations, including ethanolic extracts, aqueous extracts, fermented filtrates, and crude powder, can protect the liver against alcohol damage. Ethanol-induced ALD rats were orally treated with AC ethanolic extracts at a range of 0.25–1.25 g/kg. Hepatic damage was assessed using serum markers of liver damage, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) [84]. When liver cells are damaged, these enzymes are released into the bloodstream where they can be detected with biochemical or immunological assay kits. The AC extracts could prevent ethanol- and carbon tetrachloride (CCl4)-induced elevations of the AST, ALT, and ALP levels in the rat blood serum [85, 86], indicating that AC conferred protection against chemical-induced liver damage. Moreover, the AC ethanolic extract could normalize the alcohol-induced increase in malondialdehyde, and decreases in glutathione peroxidase, reductase, and glutathione levels in the liver [86, 87]. In the CCl4-induced ALD model, AC extracts could restore the reduction in the hepatic glutathione content and catalase (CAT) activity, and prevent free radical formation and lipid peroxidation [88]. These in vivo data were validated in a HepG2 cellular model by Kumar et al. [89], who showed that the antroquinonol from the AC ethanolic extract could protect ethanol-induced HepG2 cells against heme oxygenase 1, NF-E2 released factor (Nrf-2), and mitogen-activated protein kinase activation (MAPK).

An AC fermented filtrate protected HepG2 cells against damage caused by CCl4[90] and H2O2[91]. Song et al. [91] demonstrated that the AC fermented filtrate (0.05–0.5 mg/mL) suppressed lipid peroxidation in H2O2-induced HepG2 cells. The AC fermented filtrate restored the CCl4-induced increases in ALT, AST, lipid peroxidation, liver lesions, neutrophil infiltration, hydropic swelling, and necrosis, and the reductions in glutathione peroxidase, reductase, and transferase in animal tests [91]. The liver damage caused by chronic alcohol consumption was found to be reduced by an AC crude powder. Chen et al. [85] demonstrated that AC crude powder-treated rats had relatively smaller livers, less lipid accumulation, and lower AST, ALP, and serum alcohol levels. In addition, their serum and hepatic MMP-9 activities, and TNF-α, krueppel-like factor-6 (KLF-6), and transformation growth factor-β (TGF-β) gene expressions were downregulated. Moreover, the AC powder could enhance the metabolism of alcohol by increasing the CAT and ALDH activities [85]. Chen et al. [92] further demonstrated that the AC crude powder induced downregulation of 3-hydroxy-3-methylglutaryl-CoA reductase, sterol regulatory element-binding protein-1c, acetyl-CoA carboxylase, fatty acid synthase, and malic gene expression, and upregulation of low-density lipoprotein receptor and peroxisome proliferator-activated alpha gene expression [92]. A dried powder from fermented AC mycelium (0.34–0.57 g/kg) could protect the rat liver against damage from alcohol, and significantly suppress the alcohol-induced increases in glutamate-pyruvate aminotransferase, glutamate-oxaloacetate aminotransferase, superoxide dismutase, and CAT activities. Histological data validated the AC protective effects by showing a reduction in alcohol-induced lipid vacuole accumulation, and hydropic degradation of hepatocytes [93].

Conclusions

Hepatoprotective activities, including anti-hepatitis, anti-hepatocarcinoma, and anti-alcoholism, of AC have been demonstrated in vitro and in vivo. However, many of the underlying mechanistic actions, such as the pharmacokinetics and pharmacodynamics, remain unclear. Furthermore, human and clinical trials are required for a full evaluation of this valuable medicinal mushroom for potential clinical applications.

Abbreviations

AC:

Antrodia cinnamomea

FDA:

Food and Drug Administration

HAV:

Hepatitis A virus

HBV:

Hepatitis B virus

HCV:

Hepatitis C virus

HDV:

Hepatitis D virus

HCC:

Hepatocellular carcinoma

HBsAg:

Hepatitis B surface antigen

HBeAg:

Hepatitis B e antigen

IFN-α:

Interferon-α

WHO:

World Health Organization

IARC:

International Agency for Research on Cancer

TNF-α:

Tumor necrosis factor

IL-1β:

Interleukin-1β

IL-6:

Interleukin-6

VEGF:

Vascular endothelial cell growth factor

FGF-2:

Fibroblast growth factor-2

EC:

Endothelial cell

VEGFR-1:

VEGF receptor 1

VEGFR-2:

VEGF receptor 2

MMP:

Matrix metalloproteinase

MAA:

Methyl antcinate A

MAB:

Methyl antcinate B

TIMP-1:

Tissue inhibitor of metalloproteinases-1

TIMP-2:

Tissue inhibitor of metalloproteinases-2

ADH:

Alcohol dehydrogenase

H2O2:

Hydrogen peroxide

ALDH:

Aldehyde dehydrogenase

ALD:

Alcoholic liver disease

AST:

Aspartate aminotransferase

ALT:

Alanine aminotransferase

ALP:

Alkaline phosphatase

CCl4:

Carbon tetrachloride

CAT:

Catalase

Nrf-2:

NF-E2 released factor

MAPK:

Mitogen-activated protein kinase activation

ALDH:

Aldehyde dehydrogenase.

References

  1. Chen CJ, Su CH, Lan MH: Study on solid cultivation and bioactivity of Antrodia camphortaa. Fungal Sci. 2001, 16: 65-72.

    Google Scholar 

  2. Zang M, Su CH: Ganoderma comphoratum, a new taxon in genus Ganderma from Taiwan, China. Acta Bot Yunnanica. 1990, 12: 395-396.

    Google Scholar 

  3. Phuong do T, Ma CM, Hattori M, Jin JS: Inhibitory effects of antrodins A-E from Antrodia cinnamomea and their metabolites on hepatitis C virus protease. Phytother Res. 2009, 23: 582-584. 10.1002/ptr.2657.

    Article  PubMed  Google Scholar 

  4. Chen YS, Pan JH, Chiang BH, Lu FJ, Sheen LY: Ethanolic extracts of Antrodia cinnamomea mycelia fermented at varied times and scales have differential effects on hepatoma cells and normal primary hepatocytes. J Food Sci. 2008, 73: H179-H185. 10.1111/j.1750-3841.2008.00884.x.

    Article  CAS  PubMed  Google Scholar 

  5. Ryder SD, Beckingham IJ: ABC of diseases of liver, pancreas, and biliary system: acute hepatitis. BMJ. 2001, 322: 151-153. 10.1136/bmj.322.7279.151.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Matheny SC, Kingery JE: Hepatitis A. Am Fam Physician. 2012, 86: 1027-1034.

    PubMed  Google Scholar 

  7. Conjeevaram HS, Lok AS: Management of chronic hepatitis B. J Hepatol. 2003, 38: S90-S103.

    Article  PubMed  Google Scholar 

  8. Perrillo RP: Management of the patient with hepatitis B virus-related cirrhosis. J Hepatol. 2003, 39: S177-S180.

    Article  CAS  PubMed  Google Scholar 

  9. Chisari FV: Rous-Whipple award lecture. Viruses, immunity, and cancer: lessons from hepatitis B. Am J Pathol. 2000, 156: 1117-1132. 10.1016/S0002-9440(10)64980-2.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  10. Mulkay JP: Hepatitis B: screening and treatment. Rev Med Brux. 2012, 33: 215-222.

    CAS  PubMed  Google Scholar 

  11. Peppa D, Maini MK: Pathogenesis of hepatitis B virus infection and potential for new therapies. Br J Hosp Med (Lond). 2012, 73: 581-584.

    Article  Google Scholar 

  12. Cohen J: Report of new hepatitis virus has researchers intrigued and upset. Science. 1999, 285: 644-645. 10.1126/science.285.5428.644.

    Article  CAS  PubMed  Google Scholar 

  13. Cohen J: The scientific challenge of hepatitis C. Science. 1999, 285: 26-30. 10.1126/science.285.5424.26.

    Article  CAS  PubMed  Google Scholar 

  14. Terilli RR, Cox AL: Immunity and hepatitis C: a review. Curr HIV/AIDS Rep. 2013, 10: 51-58. 10.1007/s11904-012-0146-4.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Bouchard MJ, Navas-Martin S: Hepatitis B and C virus hepatocarcinogenesis: lessons learned and future challenges. Cancer Lett. 2011, 305: 123-143. 10.1016/j.canlet.2010.11.014.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Lee IH, Huang RL, Chen CT, Chen HC, Hsu WC, Lu MK: Antrodia camphorata polysaccharides exhibit anti-hepatitis B virus effects. FEMS Microbiol Lett. 2002, 209: 63-67. 10.1111/j.1574-6968.2002.tb11110.x.

    Article  CAS  PubMed  Google Scholar 

  17. Huang RL, Huang QL, Chen CF, Chang TT, Chou CJ: Anti-viral effects of active compounds from Antrodia camphorata on wild-type and lamivudine-resistant mutant HBV. Chin Pharmaceut J. 2003, 55: 371-379.

    CAS  Google Scholar 

  18. Popper H, Thung SN, Gerber MA: Pathology of alcoholic liver diseases. Semin Liver Dis. 1981, 1: 203-216. 10.1055/s-2008-1041748.

    Article  CAS  PubMed  Google Scholar 

  19. Schaff Z, Székely E, Járay B, Kiss A: Pathology of liver tumors. Orv Hetil. 2004, 145: 368-374.

    PubMed  Google Scholar 

  20. Parkin DM, Bray F, Ferlay J, Pisani P: Estimating the world cancer burden: globocan 2000. Int J Canc. 2001, 94: 153-156. 10.1002/ijc.1440.

    Article  CAS  Google Scholar 

  21. Parkin DM, Bray FI, Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Canc. 2001, 37: S4-S66.

    Article  Google Scholar 

  22. Chen JG, Zhu J, Parkin DM, Zhang YH, Lu JH, Zhu YR, Chen TY: Trends in the incidence of cancer in Qidong, China. 1978-2002. Int J Canc. 2006, 119: 1447-1454. 10.1002/ijc.21952.

    Article  CAS  Google Scholar 

  23. Deuffic S, Poynard T, Buffat L, Valleron AJ: Trends in primary liver cancer. Lancet. 1998, 351: 214-215.

    Article  CAS  PubMed  Google Scholar 

  24. El-Serag HB, Mason AC: Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med. 1999, 340: 745-750. 10.1056/NEJM199903113401001.

    Article  CAS  PubMed  Google Scholar 

  25. El-Serag HB, Davila JA, Petersen NJ, McGlynn KA: The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med. 2003, 139: 817-823. 10.7326/0003-4819-139-10-200311180-00009.

    Article  PubMed  Google Scholar 

  26. Taylor-Robinson SD, Foster GR, Arora S, Hargreaves S, Thomas HC: Increase in primary liver cancer in the UK, 1979-94. Lancet. 1997, 350: 1142-1143.

    Article  CAS  PubMed  Google Scholar 

  27. Wong CM, Ng IO: Molecular pathogenesis of hepatocellular carcinoma. Liver Int. 2008, 28: 160-174.

    Article  CAS  PubMed  Google Scholar 

  28. Idilman R, De Maria N, Colantoni A, Van Thiel DH: Pathogenesis of hepatitis B and C-induced hepatocellular carcinoma. J Viral Hepat. 1998, 5: 285-299. 10.1046/j.1365-2893.1998.00116.x.

    Article  CAS  PubMed  Google Scholar 

  29. WHO: Department of communable diseases. Surveillance and response: WHO. Hepatitis B. 2002

    Google Scholar 

  30. Moradpour D, Blum HE: Pathogenesis of hepatocellular carcinoma. Eur J Gastroenterol Hepatol. 2005, 17: 477-483. 10.1097/00042737-200505000-00002.

    Article  PubMed  Google Scholar 

  31. Gomaa AI, Khan SA, Toledano MB, Waked SD: Taylor-Robinson, hepatocellular carcinoma: epidemiology, risk factors and pathogenesis. World J Gastroenterol. 2008, 14: 4300-4308. 10.3748/wjg.14.4300.

    Article  PubMed Central  PubMed  Google Scholar 

  32. Farazi PA, DePinho RA: Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Canc. 2006, 6: 674-687. 10.1038/nrc1934.

    Article  CAS  Google Scholar 

  33. Bressac B, Kew M, Wands J, Ozturk M: Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa. Nature. 1991, 350: 429-431. 10.1038/350429a0.

    Article  CAS  PubMed  Google Scholar 

  34. Riley J, Mandel HG, Sinha S, Judah DJ, Neal GE: In vitro activation of the human Harvey-ras proto-oncogene by aflatoxin B1. Carcinogenesis. 1997, 18: 905-910. 10.1093/carcin/18.5.905.

    Article  CAS  PubMed  Google Scholar 

  35. Aguilar F, Harris CC, Sun T, Hollstein M, Cerutti P: Geographic variation of p53 mutational profile in nonmalignant human liver. Science. 1994, 264: 1317-1319. 10.1126/science.8191284.

    Article  CAS  PubMed  Google Scholar 

  36. Ozturk M: p53 mutation in hepatocellular carcinoma after aflatoxin exposure. Lancet. 1991, 338: 1356-1359. 10.1016/0140-6736(91)92236-U.

    Article  CAS  PubMed  Google Scholar 

  37. Hoek JB, Pastorino JG: Ethanol, oxidative stress, and cytokine-induced liver cell injury. Alcohol. 2002, 27: 63-68. 10.1016/S0741-8329(02)00215-X.

    Article  CAS  PubMed  Google Scholar 

  38. McClain CJ, Hill DB, Song Z, Deaciuc I, Barve S: Monocyte activation in alcoholic liver disease. Alcohol. 2002, 27: 53-61. 10.1016/S0741-8329(02)00212-4.

    Article  CAS  PubMed  Google Scholar 

  39. Campbell JS, Hughes SD, Gilbertson DG, Palmer TE, Holdren MS, Haran AC, Odell MM, Bauer RL, Ren HP, Haugen HS, Yeh MM, Fausto N: Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellular carcinoma. Proc Natl Acad Sci USA. 2005, 102: 3389-3394. 10.1073/pnas.0409722102.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  40. Parikh S, Hyman D: Hepatocellular cancer: a guide for the internist. Am J Med. 2007, 120: 194-202. 10.1016/j.amjmed.2006.11.020.

    Article  PubMed  Google Scholar 

  41. Ribatti D, Vacca A, Dammacco F: The role of the vascular phase in solid tumor growth: a historical review. Neoplasia. 1999, 1: 293-302. 10.1038/sj.neo.7900038.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Semela D, Dufour JF: Angiogenesis and hepatocellular carcinoma. J Hepatol. 2004, 41: 864-880. 10.1016/j.jhep.2004.09.006.

    Article  PubMed  Google Scholar 

  43. Bergers G, Benjamin LE: Tumorigenesis and the angiogenic switch. Nat Rev Canc. 2003, 3: 401-410. 10.1038/nrc1093.

    Article  CAS  Google Scholar 

  44. Carmeliet P, Jain RK: Angiogenesis in cancer and other diseases. Nature. 2000, 407: 249-257. 10.1038/35025220.

    Article  CAS  PubMed  Google Scholar 

  45. Sugimachi K, Tanaka S, Terashi T, Taguchi K, Rikimaru T, Sugimachi K: The mechanisms of angiogenesis in hepatocellular carcinoma: angiogenic switch during tumor progression. Surgery. 2002, 131: S135-S141. 10.1067/msy.2002.119365.

    Article  PubMed  Google Scholar 

  46. Hsu YL, Kuo YC, Kuo PL, Ng LT, Kuo YH, Lin CC: Apoptotic effects of extract from Antrodia camphorata fruiting bodies in human hepatocellular carcinoma cell lines. Canc Lett. 2005, 221: 77-89. 10.1016/j.canlet.2004.08.012.

    Article  CAS  Google Scholar 

  47. Hsu YL, Kuo PL, Cho CY, Ni WC, Tzeng TF, Ng LT, Kuo YH, Lin CC: Antrodia cinnamomea fruiting bodies extract suppresses the invasive potential of human liver cancer cell line PLC/PRF/5 through inhibition of nuclear factor kappaB pathway. Food Chem Toxicol. 2007, 45: 1249-1257. 10.1016/j.fct.2007.01.005.

    Article  CAS  PubMed  Google Scholar 

  48. Kuo PL, Hsu YL, Cho CY, Ng LT, Kuo YH, Lin CC: Apoptotic effects of Antrodia cinnamomea fruiting bodies extract are mediated through calcium and calpain-dependent pathways in Hep 3B cells. Food Chem Toxicol. 2006, 44: 1316-1326. 10.1016/j.fct.2006.02.009.

    Article  CAS  PubMed  Google Scholar 

  49. Hsieh YC, Rao YK, Whang-Peng J, Huang CY, Shyue SK, Hsu SL, Tzeng YM: Antcin B and its ester derivative from Antrodia camphorata induce apoptosis in hepatocellular carcinoma cells involves enhancing oxidative stress coincident with activation of intrinsic and extrinsic apoptotic pathway. J Agric Food Chem. 2011, 59: 10943-110954. 10.1021/jf202771d.

    Article  CAS  PubMed  Google Scholar 

  50. Hsieh YC, Rao YK, Wu CC, Huang CY, Geethangili M, Hsu SL, Tzeng YM: Methyl antcinate A from Antrodia camphorata induces apoptosis in human liver cancer cells through oxidant-mediated cofilin- and Bax-triggered mitochondrial pathway. Chem Res Toxicol. 2010, 23: 1256-1267. 10.1021/tx100116a.

    Article  CAS  PubMed  Google Scholar 

  51. Lee TH, Lee CK, Tsou WL, Liu SY, Kuo MT, Wen WC: A new cytotoxic agent from solid-state fermented mycelium of Antrodia camphorate. Planta Med. 2007, 73: 1412-1415. 10.1055/s-2007-990232.

    Article  PubMed  Google Scholar 

  52. Chiang PC, Lin SC, Pan SL, Kuo CH, Tsai IL, Kuo MT, Wen WC, Chen P, Guh JH: Antroquinonol displays anticancer potential against human hepatocellular carcinoma cells: a crucial role of AMPK and mTOR pathways. Biochem Pharmacol. 2010, 79: 162-171. 10.1016/j.bcp.2009.08.022.

    Article  CAS  PubMed  Google Scholar 

  53. Song TY, Hsu SL, Yen GC: Induction of apoptosis in human hepatoma cells by mycelia of Antrodia camphorata in submerged culture. J Ethnopharmacol. 2005, 100: 158-167. 10.1016/j.jep.2005.02.043.

    Article  PubMed  Google Scholar 

  54. Song TY, Hsu SL, Yeh CT, Yen GC: Mycelia from Antrodia camphorata in Submerged culture induce apoptosis of human hepatoma HepG2 cells possibly through regulation of Fas pathway. J Agric Food Chem. 2005, 53: 5559-5564. 10.1021/jf050329+.

    Article  CAS  PubMed  Google Scholar 

  55. Lin YW, Chiang BH: 4-acetylantroquinonol B isolated from Antrodia cinnamomea arrests proliferation of human hepatocellular carcinoma HepG2 cell by affecting p53, p21 and p27 levels. J Agric Food Chem. 2011, 59: 8625-8631. 10.1021/jf2011326.

    Article  CAS  PubMed  Google Scholar 

  56. Lin YW, Pan JH, Liu RH, Kuo YH, Sheen LY, Chiang BH: The 4-acetylantroquinonol B isolated from mycelium of Antrodia cinnamomea inhibits proliferation of hepatoma cells. J Sci Food Agric. 2010, 90: 1739-1744. 10.1002/jsfa.4010.

    Article  CAS  PubMed  Google Scholar 

  57. Cepeda V, Fuertes MA, Castilla J, Alonso C, Quevedo C, Pérez JM: Biochemical mechanisms of cisplatin cytotoxicity. Anticancer Agents Med Chem. 2007, 7: 3-18. 10.2174/187152007779314044.

    Article  CAS  PubMed  Google Scholar 

  58. Jordan P, Carmo-Fonseca M: Molecular mechanisms involved in cisplatin cytotoxicity. Cell Mol Life Sci. 2000, 57: 1229-1235. 10.1007/PL00000762.

    Article  CAS  PubMed  Google Scholar 

  59. Ribatti D, Vacca A, Nico B, Sansonno D, Dammacco F: Angiogenesis and anti-angiogenesis in hepatocellular carcinoma. Canc Treat Rev. 2006, 32: 437-444. 10.1016/j.ctrv.2006.06.002.

    Article  CAS  Google Scholar 

  60. Pang R, Poon RT: Angiogenesis and antiangiogenic therapy in hepatocellular carcinoma. Canc Lett. 2006, 242: 151-167. 10.1016/j.canlet.2006.01.008.

    Article  CAS  Google Scholar 

  61. Finn RS, Zhu AX: Targeting angiogenesis in hepatocellular carcinoma: focus on VEGF and bevacizumab. Expert Rev Anticancer Ther. 2009, 9: 503-509. 10.1586/era.09.6.

    Article  CAS  PubMed  Google Scholar 

  62. Chen SC, Lu MK, Cheng JJ, Wang DL: Antiangiogenic activities of polysaccharides isolated from medicinal fungi. FEMS Microbiol Lett. 2005, 249: 247-254. 10.1016/j.femsle.2005.06.033.

    Article  CAS  PubMed  Google Scholar 

  63. Yang CM, Zhou YJ, Wang RJ, Hu ML: Anti-angiogenic effects and mechanisms of polysaccharides from Antrodia cinnamomea with different molecular weights. J Ethnopharmacol. 2009, 123: 407-412. 10.1016/j.jep.2009.03.034.

    Article  CAS  PubMed  Google Scholar 

  64. Cheng JJ, Huang NK, Chang TT, Wang DL, Lu MK: Study for anti-angiogenic activities of polysaccharides isolated from Antrodia cinnamomea in endothelial cells. Life Sci. 2005, 76: 3029-3042. 10.1016/j.lfs.2004.11.023.

    Article  CAS  PubMed  Google Scholar 

  65. Chan YK, Kwok HH, Chan LS, Leung KS, Shi J, Mak NK, Wong RN, Yue PY: An indirubin derivative, E804, exhibits potent angiosuppressive activity. Biochem Pharmacol. 2012, 83: 598-607. 10.1016/j.bcp.2011.12.003.

    Article  CAS  PubMed  Google Scholar 

  66. Yue PY, Mak NK, Cheng YK, Leung KW, Ng TB, Fan DT, Yeung HW, Wong RN: Pharmacogenomics and the Yin/Yang actions of ginseng: anti-tumor, angiomodulating and steroid-like activities of ginsenosides. Chin Med. 2007, 2: 42-62.

    Article  Google Scholar 

  67. Leung KW, Yung KK, Mak NK, Yue PY, Luo HB, Cheng YK, Fan TP, Yeung HW, Ng TB, Wong RN: Angiomodulatory and neurological effects of ginsenosides. Curr Med Chem. 2007, 14: 1371-1380. 10.2174/092986707780597916.

    Article  CAS  PubMed  Google Scholar 

  68. Yue PY, Wong DY, Wu PK, Leung PY, Mak NK, Yeung HW, Liu L, Cai Z, Jiang ZH, Fan TP, Wong RN: The angiosuppressive effects of 20(R)-ginsenoside Rg3. Biochem Pharmacol. 2006, 72: 437-445. 10.1016/j.bcp.2006.04.034.

    Article  CAS  PubMed  Google Scholar 

  69. Fan TP, Yeh JC, Leung KW, Yue PY, Wong RN: Angiogenesis: from plants to blood vessels. Trends Pharmacol Sci. 2006, 27: 297-309. 10.1016/j.tips.2006.04.006.

    Article  CAS  PubMed  Google Scholar 

  70. Kok TW, Yue PY, Mak NK, Fan TP, Liu L, Wong RN: The anti-angiogenic effect of sinomenine. Angiogenesis. 2005, 8: 3-12. 10.1007/s10456-005-2892-z.

    Article  CAS  PubMed  Google Scholar 

  71. Charels K, Klöppel G: Pathology and pathogenesis of toxic (alcoholic) liver disease. Hepatogastroenterology. 1991, 38: 189-194.

    CAS  PubMed  Google Scholar 

  72. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Personal habits and indoor combustions. Volume 100 E. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum. 2012, 100: 1-538.

    Google Scholar 

  73. Yip WW, Burt AD: Alcoholic liver disease. Semin Diagn Pathol. 2006, 23: 149-160. 10.1053/j.semdp.2006.11.002.

    Article  PubMed  Google Scholar 

  74. Doria JJ: Alcohol metabolism. Alcohol Health Res World. 1997, 21: 323-

    Google Scholar 

  75. Bode C, Bode JC: Alcohol absorption, metabolism, and production in the gastrointestinal tract. Alcohol Health Res World. 1997, 21: 82-83.

    Google Scholar 

  76. Zakhari S: Overview: How is alcohol metabolized by the body?. Alcohol Res Health. 2006, 29: 245-254.

    PubMed  Google Scholar 

  77. Lieber CS: Hepatic, metabolic and toxic effects of ethanol: 1991 update. Alcohol Clin Exp Res. 1991, 15: 573-592. 10.1111/j.1530-0277.1991.tb00563.x.

    Article  CAS  PubMed  Google Scholar 

  78. Lieber C: The discovery of the microsomal ethanol oxidizing system and its physiologic and pathologic role. Drug Metab Rev. 2004, 36: 511-529. 10.1081/DMR-200033441.

    Article  CAS  PubMed  Google Scholar 

  79. Donohue TM: Alcohol-induced steatosis in liver cells. World J Gastroenterol. 2007, 13: 4974-4978.

    CAS  PubMed  Google Scholar 

  80. Sozio M, Crabb DW: Alcohol and lipid metabolism. Am J Physiol Endocrinol Metab. 2008, 295: E10-E16. 10.1152/ajpendo.00011.2008.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  81. Bruha R, Dvorak K, Petrtyl J: Alcoholic liver disease. World J Hepatol. 2012, 4: 81-90. 10.4254/wjh.v4.i3.81.

    Article  PubMed Central  PubMed  Google Scholar 

  82. Syn WK, Teaberry V, Choi SS, Diehl AM: Similarity and differences in the pathogenesis of alcoholic and non-alcoholic steatohepatiotis. Semin Liver Dis. 2009, 29: 200-210. 10.1055/s-0029-1214375.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  83. Fernández-Checa JC, Kaplowitz N, Colell A, García-Ruiz C: Oxidative stress and alcoholic liver disease. Alcohol Health Res World. 1997, 21: 321-324.

    PubMed  Google Scholar 

  84. Lieber CS: Alcohol, liver, and nutrition. J Am Coll Nutr. 1991, 10: 602-632. 10.1080/07315724.1991.10718182.

    Article  CAS  PubMed  Google Scholar 

  85. Wu MT, Tzang BS, Chang YY, Chiu CH, Kang WY, Huang CH, Chen YC: Effects of Antrodia camphorata on alcohol clearance and antifibrosis in livers of rats continuously fed alcohol. J Agric Food Chem. 2011, 59: 4248-4254. 10.1021/jf104561h.

    Article  CAS  PubMed  Google Scholar 

  86. Lu ZM, Tao WY, Zou XL, Fu HZ, Ao ZH: Protective effects of mycelia of Antrodia camphorata and Armillariella tabescens in submerged culture against ethanol-induced hepatic toxicity in rats. J Ethnopharmacol. 2007, 110: 160-164. 10.1016/j.jep.2006.09.029.

    Article  CAS  PubMed  Google Scholar 

  87. Lu ZM, Tao WY, Xu HY, Ao ZH, Zhang XM, Xu ZH: Further studies on the hepatoprotective effect of Antrodia camphorata in submerged culture on ethanol-induced acute liver injury in rats. Nat Prod Res. 2011, 25: 684-695. 10.1080/14786410802525487.

    Article  CAS  PubMed  Google Scholar 

  88. Hsiao G, Shen MY, Lin KH, Lan MH, Wu LY, Chou DS, Lin CH, Su CH, Sheu JR: Antioxidative and hepatoprotective effects of Antrodia camphorata extract. J Agric Food Chem. 2003, 51: 3302-3308. 10.1021/jf021159t.

    Article  CAS  PubMed  Google Scholar 

  89. Kumar KJ, Chu FH, Hsieh HW, Liao JW, Li WH, Lin JC, Shaw JF, Wang SY: Antroquinonol from ethanolic extract of mycelium of Antrodia cinnamomea protects hepatic cells from ethanol-induced oxidative stress through Nrf-2 activation. J Ethnopharmacol. 2011, 136: 168-177. 10.1016/j.jep.2011.04.030.

    Article  CAS  PubMed  Google Scholar 

  90. Lin WC, Kuo SC, Lin WL, Fang HL, Wang BC: Filtrate of fermented mycelia from Antrodia camphorata reduces liver fibrosis induced by carbon tetrachloride in rats. World J Gastroenterol. 2006, 12: 2369-2374.

    PubMed Central  CAS  PubMed  Google Scholar 

  91. Song TY, Yen GC: Protective effects of fermented filtrate from Antrodia camphorata in submerged culture against CCl4-induced hepatic toxicity in rats. J Agric Food Chem. 2003, 51: 1571-1577. 10.1021/jf0209701.

    Article  CAS  PubMed  Google Scholar 

  92. Huang CH, Chang YY, Liu CW, Kang WY, Lin YL, Chang HC, Chen YC: Fruiting body of Niuchangchih (Antrodia camphorata) protects livers against chronic alcohol consumption damage. J Agric Food Chem. 2010, 58: 3859-3866. 10.1021/jf100530c.

    Article  CAS  PubMed  Google Scholar 

  93. Dai YY, Chuang CH, Tsai CC, Sio HM, Huang SC, Chen JC, Hu ML: The proliferation of Anthrodia camphorate against acute hepatotoxicity of alcohol in rat. J Food Drug Ann. 2003, 11: 177-185.

    Google Scholar 

Download references

Acknowledgments

K.S.Y. Leung would like to thank the Hong Kong Baptist University for financial support. The generous support of GeneFerm Biotechnology Company Limited of Taiwan for the entire research on Antrodia cinnamomea is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to PatrickYing-Kit Yue or KelvinSze-Yin Leung.

Additional information

Competing interests

The authors declare that they have no competing interest.

Authors' contributions

PYKY was responsible for drafting and proofreading of this article. The information searching was done by YYW, KYKW, and YKT. Final proofreading was done by KSYL. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Yue, PK., Wong, YY., Wong, KK. et al. Current evidence for the hepatoprotective activities of the medicinal mushroom Antrodia cinnamomea. Chin Med 8, 21 (2013). https://doi.org/10.1186/1749-8546-8-21

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1749-8546-8-21

Keywords