search
for
 About Bioline  All Journals  Testimonials  Membership  News


African Health Sciences
Makerere University Medical School
ISSN: 1680-6905 EISSN: 1729-0503
Vol. 10, Num. 1, 2010, pp. 93-98

African Health Sciences, Vol. 10, No. 1, March, 2010, pp. 93-98

Hepatocellular carcinoma and the underlying mechanisms

*Oyagbemi AA, Azeez OI, Saba AB

Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Oyo State, Nigeria.

*Corresponding author: Oyagbemi A.A, Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Oyo State, Nigeria. E-mail: ademolaoyagbemi@yahoo.com , aa.oyagbemi@mail.ui.edu.ng Phone: +2348033639776 Fax: 02-8103043

Code Number: hs10017

Abstract

The incidence of hepatocellular carcinoma is increasing worldwide as well as the associated risk factors, some of which include exposure to aflatoxin B1, Hepatitis B (HBV) virus and hepatitis C (HCV) virus.  Mutation of tumour suppressor gene p53 at codon 249ser at exon 7 has been found to contribute significantly to replication of damaged DNA and subsequent tumour progression. The x gene of HBV (HBx) is the most common open reading frame integrated into the host genome in hepatocellular carcinoma and the integrated HBx is frequently mutated in hepatocellular carcinoma. Mutant HBx proteins still retain their ability to bind to p53 thereby attenuating DNA repair and p53-mediated apoptosis.

Keywords: hepatocellular carcinoma, aflatoxin B1, HBV, HCV, p53

Introduction

Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. It is the fourth leading cause of cancer-related death in the world.1 The major risk factors include chronic infections with the hepatitis B (HBV) or C (HCV) virus and exposure to dietary AFB1 or alcohol consumption. A link based on circumstantial evidence has been divulged between high exposure to AFBI and mutation at the 3rd nucleotide base of codon 249, which is located on the 7th exon of p53 gene of cells of primary liver cancer from patients in tropical countries of the world and activation of the WNT signal transduction pathway.2-6 AFB1 frequently induces G: C to T: A transversions at the third base in codon 249. Interestingly, mutant DNA in plasma is a biomarker of both AFBI exposure and potential risk factor for HCC with subsequent p53 mutation.7 The tumour suppressor gene p53 is the most commonly mutated gene in human cancers. 8

Chronic infections with HBV and HCV viruses and oxyradical disorders including hemochromatosis also generate reactive oxygen/nitrogen species that both damage DNA and mutate cancer - related genes such as tumour suppressor gene p53.9 The p53 biological network is a key responder to this oxidative and nitrosative stress. Depending on the extent of the DNA damage, p53 regulate transcription of protective antioxidant genes and the extent of DNA damage that ultimately trans-activates pro-oxidant genes which eventually contribute to apoptosis. The x gene of HBV (HBx) is the most common open reading frame integrated into the host genome in HCC and the integrated HBx is frequently mutated. Mutant HBx proteins still retain their ability to bind to p53 and attenuate DNA repair and p53-mediated apoptosis. Hence, both viruses and chemicals (especially vinyl chloride) are implicated in the etiology of p53 mutation during the molecular pathogenesis of HCC.

HCC is a major cause of cancer morbidity and mortality in many parts of the world, including Asia and Sub Saharan Africa, where there are >500,000 new cases each year and >200,000 deaths annually in the People's Republic of China (P.R.C) alone.10 The major etiological factors associated with development of HCC in these regions are infection with HBV and or HCV and long time exposure to high levels of AFBI in the diet.11-12

Mechanisms underlying Hepatocarcinogenesis

The biology, mode of transmission, and epidemiology of HBV continue to be actively investigated and have been recently reviewed.13 A mutation in the HBV genome can alter the expression of multiple proteins. In many cases of HCC in China and Africa, a double mutation in the HBV genome, an adenine-to-thymine transversion at nucleotide 1762 and a guanine-to-adenine transition at nucleotide 1764 (1762T/1764A) has been found in tumours.14-15

This segment of the HBV genome contains an overlapping sequence for the base core promoter region and the HBx gene; therefore, the double mutation in codon 130 and 131 of the HBx gene reported in human HCC is identical to the 1762 and 1764 nucleotide changes.16 The onset of these mutations was shown to be associated with the increasing severity of the HBV infection and cirrhosis.14-15 HBx in transformed hepatocyte has been demonstrated to inhibit the repair of damaged hepatocyte DNA. This effect may be mediated by interaction with p53 or through binding to the damaged DNA binding protein (DDB), which plays an accessory role in nucleotide excision repair.13 In addition, HBx activates cell signalling cascades involving mitogen-activated protein kinase (MAPK) and Janus family tyrosine kinases (JAK)/signal transducer and activators of transcription (STAT) pathways.13 The process by which tumour DNA is released into circulating blood is unclear but may result from accelerated necrosis,apoptosis, or other processes.17 A specific codon 249 p53 mutation detectable in plasma samples at the time of HCC diagnosis, can be measured in some individual at least 5 years before diagnosis.18

Heterogeneity in etiological factors of HCC

The frequency of HCC is particularly high in Asia and Africa due to the high frequency of viral hepatitis infections and to Aflatoxin B1 exposure (AFB1). Over the last 10 years, the incidence of HCC has noticeably increased in United Kingdom, France and United States. This is probably linked to viral hepatitis C infections. Etiological factors that are associated with the development of hepatic tumours are well known in these regions. They include infection with the hepatitis B virus (HBV) or hepatitis C virus (HCV), heavy alcohol intake, prolonged dietary exposure to AFBl or vinyl chloride and primary hemochromatosis. In 90% of the HCC cases, at least one of these risk factors can be identified either alone or in combination with another factor. The presence of each risk factor among patients varies according to the geographical origin of the patients. Globally, exposure to HCV, HBV and AFBI are responsible for about 80% of all HCC in humans' worldwide but the principal risk factor varies between countries. In Japan almost all HCC are linked to HCV infection, whereas in Africa HBV infections are predominant.19 In France, HBV and HCV infections and alcohol intake are identified with approximately equal frequency. Exposure to AFBI is commonly found in sub-tropical countries where humid heat can lead to the development" of Aspergillus flavus in improperly stored foods such as cereals and peanuts. This mycotoxin is strongly hepatocarcinogenic in experimental animal models and acts synergistically with HBV infection to increase the risk of HCC.20 Tobacco exposure is the leading carcinogen associated with multiple solid tumours21. Several investigators have previously reported an association between tobacco and HCC with odds ratios ranging from 1.5 to 6.8.22-23 However, other studies found no association between tobacco and HCC.24

Hepatocarcinogenesis

The different risk factors of HCC include chronic lesions in the liver with associated inflammation, necrosis of hepatocytes and fibrosis. Overall, HCC development is closely associated with cirrhosis and more than 80% of the tumours are found in a chronic hepatitis or a cirrhotic background.25 Dysplastic nodules and macroregenerative nodules have long been considered to be the likely precursors of HCC because of their frequent association with the HCC occurrence.26 Chromosome aberrations occur in HCC and these may already contain genetic aberrations. However, in rare cases (less than 10% of the cases), HCC are observed in non-cirrhotic liver and even without inflammatory lesions. The HCC which develop in an otherwise normal liver are usually found in patients without well-established risk factors. Some of these cases may correspond to the malignant transformation of liver adenoma that are rare benign hepatocellulartumours sometimes found in young women taking oral contraceptives.27

HBV infection

The incidence of HCC has been shown to vary widely worldwide. 28 Among males, the highest incidence rates are found in eastern Asia, particularly in China where HCC was reported to be the third most common cause of cancer death29. Chronic infection with the HBV has been reported by various authors as the strongest risk factor for HCC worldwide. 28, 30-32 However, populations with similar prevalence of HBV infection have different incidence of HCC, suggesting the presence of other important risk factors. Aflatoxins, a group of mycotoxins produced by the common fungi Aspergillus flavus and Aspergillus parasiticus, are established human hepatocarcinogens and are well-known HCC risk factors when present in foodstuffs.32-35 Some epidemiological and animal studies have found evidence for an HBV-aflatoxin interaction in hepatocarcinogenesis. 35-41

Several mechanisms underlying this principle have been proposed to explain the interaction between HBV and aflatoxin. The increase in cellular proliferation induced by HBV could increase the probability for clonal expansion of an existing aflatoxin induced-p53 249ser mutation42. An increase in levels of aflatoxin metabolism enzymes (e.g., P450 enzymes in which its activity is associated with increased hepatotoxicity of aflatoxin) has been described for HBV transgenic mice and has been postulated as a mechanism for interaction.43 The HBx protein, which is encoded by HBV interferes with the nucleotide excision repair pathway,  a major repair pathway which cells use to repair damaged DNA.44 However, the presence of mutant HBx protein could increase the frequency of aflatoxin-induced mutations.44 Also, HBV infection was reported to increase oxidative stress, which could lead to an increase in p53 mutations.45

Mechanisms of HBV-mediated hepatocarcinogenesis

HBV infection can promote carcinogenesis by at least 3 different mechanisms. First, integration of the viral DNA in the host genome can induce chromosome instability. Second, insertional mutations of HBV are known to activate endogenous genes of retinoic acid α-receptor, cyclin A and mevalonate kinase which are involved in cell cycle control, cellular proliferation and differentiation. The second mechanism is associated with specific intracellular receptors. Recently, 15 new genes were found to be altered by HBV integration in tumors suggesting that viral integration in the vicinity of genes controlling cell proliferation, viability and differentiation is a mechanism frequently involvedin HBV hepatocarcinogenesis. The third mechanism of carcinogenesis linked to HBV infection is based on the expression of viral protein, in particular HBx, to modulate cell proliferation and viability. Moreover, HBx binds to p53 and inactivates p53-dependent activities, including p53-mediated apoptosis. Recently, the association between hepatitis B virus and Hepatocellular carcinoma and the molecular mechanism of action that is involved in the hepatocarcinogenesis has been extensively described.46,47

Interaction of AFB1 with DNA and chromatin proteins (histone)

After an exposure to AFB1, accumulations of damaged DNA are found in the liver, as a result of conversion of the AFBI to its active metabolites. AFBI is a very potent mutagen and the AFBI epoxides (active metabolites of aflatoxin) react with guanine in DNA, leading to genetic changes. The most frequent mutation induced is the (guanine-cytosine to thymine-adenine) GC to TA transversion. However, quantitative determination of AFB1 in human aflatoxin albumin adducts has been ellucidated.48 The mutational pattern of p53 gene in HCC from regions where AFBI exposure level is high, revealed (guanine to thymine) G to T transversion at codon 249 in more than 50% of the cases. A more detail study revealed that AFB1 binds preferentially to lysyl amino acid residues in histone proteins.10 The binding of AFB1 to histone proteins has significant functional implications because histone has been reported to be the packaging material for DNA and histone H1 is the most external of the histone proteins wrapped around DNA.10,49 Because of the high content of basic amino acids in histones, it is conjectured that there is a strong electrostatic interaction between them and DNA and that (addition of acetyl group) acetylation of the lysyl sites which is involved in this type of interaction, reduces the net positive charge of the histone and loosens the bonds between histone and DNA. Acetylation is reported to occur at the amino group of lysly amino acid residues which is the same binding site of AFBl.50, 10

The effect of AFBl binding to histone is therefore likely to be similar to reaction elicited by Acetylation (a post transcriptional modification), which is the partial loosening of the histone-DNA bond and the consequent degradation of the histone by specific proteases.51It is generally accepted that such a partial loosening of the histone DNA bonds always precedes gene expression. This means that it is most likely that it is the binding of AFBl to lysly amino acid residues in histone with the consequent loosening of the histone-DNA bond that makes p53 accessible for damage. It is also likely that the binding of AFB1 to histones with the consequent loosening of histone is primary to its binding to the DNA of p53 genes even though the binding to DNA subsequently exceeds its binding to histone. Taken together, the binding of AFBI to DNA is responsible for the inhibition of RNA Synthesis, which is involved in gene expression.

The implication of the above is that it is the binding to chromatin proteins (histone) may be involved in the expression of the mutated p53 gene resulting from the interaction of AFBI with DNA and chromatin proteins. The p53 gene is reported to be mutated in HCC after exposure to aflatoxin.52 Recently, some authors have extensively discussed the association between AFB1 and the associated risk factors involved in HCC.53-55

Conclusion

The nexus between hepatocellular carcinoma and the associated risk factors cannot be overemphasized. The interaction between aflatoxin and HBV or HCV in hepatocarcinogenesis and multi-stage carcinogenesis is grossly elucidated. Characterization of the genetic alterations associated with HCC tumors is an essential step to increase our knowledge of hepatocarcinogenesis. Systematic search for these alterations in series of tumors including tumour grades, stages, etiologies and the associated pre-neoplastic lesions is therefore necessary to find and identify the pattern of accumulation of the genetic alterations during tumour progression. Microarray analysis and metagenomics may also contribute significantly to identifying new carcinogenetic pathways altered in these tumors. New insight should therefore be geared towards getting a better clinical application, to identity tumour markers that are useful for early detection of tumors, to predict prognosis, or to find new therapeutic targets with their underlying molecular mechanism of action.

References

  1. Murugavel KG, Naranatt PP, Shankar et al.: Prevalence of aflatoxin B1 in liver biopsies of prove hepatocellular carcinoma in India determined by an in-house immunoperoxidase  test. J Med Microbiol 2007; 56(Pt 11):1455 PubMed -9.
  2. Mariana  CS, David  MU, Mimi  CYu et al.: Hepatitis B, Aflatoxin B1, and p53 Codon 249 Mutation in Hepatocellular Carcinomas from Guangxi, People's Republic of China,  and a Meta-analysis of Existing Studies. Cancer Epidemiology Biomarkers & Prevention  2001; 10: 617-625.
  3. Bressac B, Kew M, Wands J, Ozturk M: Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa. Nature (Lond.), 1991; 350: 429-431.        
  4. Hsu IC, Metcalf RA, Sun T et al.: Mutational hotspot in the p53 gene in human  hepatocellular carcinomas. Nature (Lond.). 1991; 350: 427- 428.
  5. Heyward N, Walder G, Graham W, Cooksley, E. Hepatocellular carcinoma  mutation (Letter) Nature (Lond.) 1996; 350: 427-428.
  6. Ozturk M, Bressae B, Puisieux et al.: P53 mutation in hepatocellular carcinoma after aflatoxin exposure. Lancet. 1991; 338: 113-122.
  7. Shuang YK,  Suree  L,  Niwat  et al.:Hepatitis B 1762T/1764A Mutations, Hepatitis C Infection, and Codon 249 p53 Mutations in Hepatocellular Carcinomas from Thailand.  Cancer Epidemiology Biomarkers & Prevention 2005; 14: 380-384.
  8. Greenblatt  MS, Bennett W P, Hollstein M, Harris C C: Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 1994; 54: 48554878.
  9. Paradis V, Mathurin P, Kollinger M et al.: In situ detection of lipid peroxidation of Chronic hepatitis C: correlation with pathological features. J Clin Pathol 1997; 50: 401 406.
  10. Li ND, Lu FZ, Zhang SW, Mo R, Sun XD: Trends and prediction in malignant tumors mortality in past 20 years in China.. Chin. J Oncol 1997; 19: 3-9.
  11. Qian GS, Yu MC, Ross et al.: DA follow-up study of urinary markers of aflatoxin exposure and liver cancer risk in Shanghai, P.R.C. Cancer Epidemiol  Biomark Prev 1994; 3: 3-11.
  12. Kensler TW, Qian GS, Chen JG, Groopman  JD: Hepatocarcinogensis as a paradigm for cancer prevention. Nat Rev Cancer 2003; 3:32129.
  13. Arbuthnot P, Capovilla A, Kew M: Putative role of hepatitis B virus X protein in hepatocarcinogenesis: effects on apoptosis, DNA repair, mitogen-activated protein kinase and JAK/STAT pathways. J Gastroenterol Hepatol 2000; 15(4):339-41
  14. Hsu IC, Metcalf RA, Sun T, Welsh  JA, Wang  NJ, Harris  CC: Mutational hotspot in  the p53 gene in human hepatocellular carcinomas. Nature (Lond.), 1991; 350: 427-428.
  15. Baptista  M, Kramvis  A, Kew  MC: High prevalence of 1762(T) 1764(A) mutations in the basic core promoter of hepatitis B virus isolated from black Africans with hepatocellular carcinoma compared with asymptomatic carriers. Hepatology 1999;         29:94653.
  16. Zhang F, Zhu Y, Sun Z: Universal presence of HBVx gene and its close association  with hotspot mutation of p53 gene in hepatocellular carcinoma of prevalent area in China. Zhonghua Zhongliu Zazhi 1998; 20:18-21.
  17. Anker P, Mulcahy H, Chen  XQ, Stroun  M: Detection of circulating tumor DNA in the blood (plasma/serum) of cancer patients. Cancer Metastasis Rev. 1999; 18:6573.
  18. Jackson PE, Kuang  SY, Wang  JBet al.: Prospective detection of codon 249 mutations in plasma of hepatocellular carcinoma patients. Carcinogenesis (2003; 24:17.
  19. Bosch FX, Ribes J, Díaz M, Cléries R:  Primary liver cancer: worldwide incidence and  trends. Gastroenterology 2004; 127(5 Suppl PubMed  1):S5-S16.
  20. Chen CJ, Wang LY, Lu et al.Elevated aflatoxin exposure and Increased risk of Hepatocellular carcinoma. Hepatology 1996; 24:38-42.
  21. Adami HO, Hsing AW, McLaughlin et al:. Alcoholism and liver cirrhosis in the etiology of primary liver cancer. Int J Cancer 1992; 51:898-902.
  22. Hadzlyannis S, Tabor E, Kaklamani et al:. A case-control study of hepatitis Band C virus infections in the etiology of hepatocellular carcinoma. Int J Cancer 1995; 60:627 -631. 
  23. Tzonou  A, Trichopoulos  D, Kaklamani  E et al.: Epidemiologic assessment of Interactions of hepatitis-C virus with seromarkers of hepatitis-B and D viruses, cirrhosis and tobacco smoking in hepatocellular carcinoma. Int  J Cancer 1991; 377-380.
  24. Austin H, Deizell E, Grufferman et al:. A case-control study of Hepatocellular carcinoma and the hepatitis B virus, cigarette smoking and alcohol  consumption. Cancer Res 1986; 46:962-966.
  25. Yu MW, Hsu FC, Sheen IS et al.: Prospective Study of Hepatocellular Carcinoma and Liver Cirrhosis in Asymptomatic Chronic Hepatitis B Virus Carriers. American Journal of  Epidemiology 1997; 145(11): 1039 PubMed -1047.
  26. Terada T, Veda K, Nakanuma Y: Histopathological and morphometric analysis of atypical adenomatous hyperplasia of human cirrhotic livers. Virchows Arch A Pathol Anat Histopathol 1993; 422:381-8.
  27. Ham JM, Stevenson D, Liddelow AG: Hepatocellular carcinoma possibly induced by oral contraceptives. Digestive Diseases and Sciences 2005; 1575-2568.  
  28. Yeh F-S, Yu MC, Mo C-C, Tong MJ, Henderson BE: Hepatitis B virus, aflatoxins, and hepatocellular carcinoma in Southern Guangxi, China.  Cancer Res 1989; 49: 25062509.
  29. National Cancer Control Office: Nanjing Institute of Geography. Atlas of Cancer Mortality in the People's Republic of China. Shanghai: China Map Press. 1979.
  30. Blumberg BS, and London WT: Hepatitis B virus and the prevention of primary cancer of the liver. J Natl Cancer Inst (Bethesda) 1985; 74: 267273.
  31. London WT, Evans AA, Buetow  et al.: Molecular and genetic epidemiology of  hepatocellular  carcinoma: studies in China and Senegal. Princess Takamatsu Symp. 1995; 25: 5160.
  32. Vainio H, Heseltine E, Wilbourn J: Report on an IARC working group meeting on some naturally occurring substances. Int J Cancer 1993; 53: 535537.
  33. Busby WF, and Wogan GN: Aflatoxins. In: C. E. Searle (ed). Chemical Carcinogens 1984; 2:9451136 PubMed . Washington D. C.: American Chemical Society
  34. Groopman  JD: Do aflatoxin-DNA adduct measurements in humans provide  accurate data for cancer risk assessment? IARC Sci Publ 1988; 89: 5562.
  35. Qian GS, Yu MC, Ross  et al.: A follow-up study of urinary markers of aflatoxin exposure and liver cancer risk in Shanghai, P.R.C Cancer Epidemiol  Biomark Prev  1994; 3:3-11.
  36. Lunn RM, Zhang Y-J, Wang et al:. P53 mutations, chronic hepatitis B virus infection, and aflatoxin exposure in hepatocellular carcinoma in Taiwan. Cancer Res 1997; 57: 3471 3477.
  37. Ross RK, Yuan JM, Yu et al.: Urinary aflatoxin biomarkers and risk of hepatocellular carcinoma. Lancet  1992; 339: 943946.
  38. McGlynn KA, Rosvold EA, Lustbader et al:. Susceptibility to Hepatocellular carcinoma is associated with genetic variation in the enzymatic detoxification of aflatoxin B1. Proc Natl  Acad  Sci USA  1995; 92: 23842387.
  39. Dragani TA, Manenti G, Farza H, Della-Porta G, Tiollais P, and Pourcel C. Transgenic mice containing hepatitis B virus sequences are more susceptible to carcinogen-induced hepatocarcinogenesis. Carcinogenesis (Lond.) 1990; 11: 953956.
  40. Sell S, Hunt JM, Dunsford HA, Chisari FV: Synergy between hepatitis B virus expression and chemical hepatocarcinogens in transgenic mice. Cancer Res 1991; 51: 12781285.
  41. Montesano R, Hainaut P, and Wild CP: Hepatocellular carcinoma: from gene to public health. J. Natl. Cancer Inst. (Bethesda) 1997; 89: 18441851.
  42. Chisari FV, Klopchin K, Moriyama et al.: Molecular pathogenesis of hepatocellular carcinoma in hepatitis B virus transgenic mice. Cell 1989; 59: 11451156.
  43. Kirby GM, Chemin I, Montesano R, Chisari FV, Lang MA, and Wild CP: Induction of specific cytochrome P450s involved in aflatoxin B metabolism in hepatitis B virus transgenic mice. Mol  Carcinog  1994; 11: 7480
  44. Jia L, Wang X W, and Harris CC: Hepatitis B virus, X protein inhibits nucleotide excision repair. Int  J  Cancer 1999; 80: 875879.
  45. Hussain S P, Aguilar F, Amstad P, and Cerutti P: Oxy-radical induced mutagenesis of hotspot codons 248 and 249 of the human p53 gene. Oncogene 1994; 9: 22772281.
  46. Liu  ZM, Li  LQ, Peng  et al:. Hepatitis B virus infection contributes to oxidative stress  in a population exposed to aflatoxin B1 and high-risk for hepatocellular carcinoma. Cancer Lett 2008; 263(2):212-22.
  47. Wu HC, Wang Q, Yang et al.: Urinary 15-F2t-isoprostane, aflatoxin B1 exposure and  hepatitis B virus infection and hepatocellular carcinoma in Taiwan. Carcinogenesis. 2008; 29(5):971-6.
  48. McCoy LF, Scholl  PF, Sutcliffe  et al.: Human aflatoxin albumin adducts quantitatively  compared by ELISA, HPLC with fluorescence detection, and HPLC with isotope dilution   mass spectrometry. Cancer Epidemiol Biomarkers Prev (2008; 17(7):1653-7.
  49. Stelolwagen RH, and Cole RD: Chromosomal proteins Ann Rev Biochem 1969; 38: 951.
  50. Sung MT, and Dixon GH. Modifications of histones during spermiogenesis in trout: A molecular mechanism or altering histone binding .to DNA proc. Natll. Acad. Sci U.S.A. 1970; 67: 1616.
  51. Candido EPM, and Dixon GH. Trout testis cells: Acetvlation of histones in different cells types from developing trouts tests. J  BioI Chem 1972; 247: 5506.
  52. Paget V, Sichel  F, Garon  D, Lechevrel M:  Aflatoxin B1-induced TP53 mutational   pattern in normal human cells using the FASAY (Functional Analysis of Separated   Alleles in Yeast). Mutat Res 2008; 656(1-2):55-61.
  53. Abdel-Wahab M, Mostafa  M, Sabry  M, el-Farrash  M, Yousef T: Aflatoxin as a risk  factor for hepatocellular carcinoma in Egypt, Mansoura Gastroenterology Center study. Hepatogastroenterology  2008; 55(86-87):1754-9.
  54. Murugavel KG, Naranatt PP, Shankar et al:. Prevalence of aflatoxin B1 in liver biopsies of proven hepatocellular carcinoma in India determined by an in-house  immunoperoxidase test. J Med Microbiol 2007; 56(Pt 11):1455 PubMed -9.
  55. Long XD, Ma Y, Qu de  et al.:The polymorphism of XRCC3 codon 241 and AFB1-  related hepatocellular carcinoma in Guangxi population, China. Ann Epidemiol 2008; 18(7):572- 8.

Copyright © 2010 - Makerere Medical School, Uganda

Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil