Mitochondrial DNA haplogroup M is associated with late onset of hepatocellular carcinoma

  • Authors:
    • Zhanjun Guo
    • Hua Yang
    • Cuiju Wang
    • Shufeng Liu
  • View Affiliations

  • Published online on: December 27, 2011     https://doi.org/10.3892/etm.2011.434
  • Pages: 499-502
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The accumulation of single nucleotide polymorphisms (SNPs) in the displacement loop (D-loop) of mitochondrial DNA (mtDNA) has been associated with various types of cancer. The association of SNPs with cancer risk and disease outcome has been exhaustively studied. In this study, we investigated the association of age-at-onset and SNPs in the mitochondrial D-loop using a population-based series of hepatocellular carcinoma (HCC) patients. Haplogroup M (489C) and allele 235G were identified for their association with the late onset of HCC by the log-rank test. In an overall multivariate analysis, haplogroup M (489C) was identified as an independent predictive factor for the age-at-onset of HCC at borderline significant levels [relative risk, 1.736; 95% confidence interval (CI), 0.967-3.115; p=0.065]. Genetic polymorphisms in the D-loop are predictive markers for age‑at-onset in HCC patients. Accordingly, the analysis of genetic polymorphisms in the mitochondrial D-loop may help to identify HCC patient subgroups at high risk of early onset of the disease.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most frequent type of cancer and the third leading cause of cancer mortality worldwide, with over half a million cases of mortality every year (1). HCC is also common in China. According to the annual cancer incidence and mortality report, the incidence and mortality rates of HCC in China over the last decade were 300,000 and 306,000 cases, respectively (2,3). This disease is strongly associated with several risk factors, including chronic hepatitis B virus (HBV) and chronic hepatitis C virus (HCV) infection, as well as alcohol abuse (4). Certain epidemic factors have been identified as risk factors or outcome predictors for HCC (57); however, the true mechanism of this cancer remains unknown. To date, few studies have focused on the genetic factors associated with age-at-onset of this cancer, although they have demonstrated the genetic prevalence of this disease (8).

Hepatitis virus infection and alcohol abuse are associated with increased oxidative stress in liver cells, resulting in DNA changes including mitochondrial DNA (mtDNA) instability (9,10). The human mitochondrial genome is 16 kb in length and is a closed-circular duplex molecule that contains 37 genes, including two ribosomal RNAs and complete sets of 22 transfer RNAs (tRNAs) (11). mtDNA is believed to be more susceptible to DNA damage and acquires mutations at a higher rate than nuclear DNA owing to high levels of reactive oxygen species (ROS), lack of protective histones and limited capacity for DNA repair in the mitochondria (1214). Thus, somatic mtDNA mutations occur in a wide variety of degenerative diseases and cancers (15,16) and may be homoplasmic by clonal expansion (17,18) or heteroplasmic in tumor tissues (19,20). In a number of cancers, including hepatitis virus-related HCC, somatic mutations are frequently located in the mtDNA non-coding region, termed the displacement loop (D-loop) (21,22). This region is important for regulating the replication and expression of the mitochondrial genome since it contains the leading-strand origin of replication and is the main promoter for transcription (23).

We sequenced the D-loop that contains a length of 1,122 bps (nucleotides 16024-16569 and 1–576; www.mitomap.org) in the blood from HCC patients and identified 92 single nucleotide polymorphims (SNPs) in the D-loop. We also identified cancer risk and outcome associated SNPs (24,25). In the present study, we assess the correlation between germline SNPs of the D-loop and age-at-onset in HCC patients.

Materials and methods

Tissue specimens and DNA extraction

Blood samples were collected at the Fourth Hospital of Hebei Medical University (China) from 60 HCC patients who underwent HCC resection in the Department of Hepatobiliary Surgery between 2007 and 2008. All patients originated from the Hebei Province of China, a high-risk area for HCC. Whole blood was obtained from corresponding HCC patients. Mitochondria isolation and mtDNA extraction were carried out using the Blood Mitochondrial DNA Extraction Kit (Genmed Scientific Inc., Shanghai, China). The study was approved by the Human Tissue Research Committee of the Fourth Hospital of Hebei Medical University. All patients provided written informed consent for the collection of samples and subsequent analysis.

Polymerase chain reaction (PCR) amplification and sequence analysis

The forward primer, 5′-CCCCATGCTTACAAGCAA GT-3′ (nucleotide 16190–16209); and reverse primer, 5′-GCTTT GAGGAGGTAAGCTAC-3′ (nucleotide 602-583) were used for the amplification of a 982-bp product from the mtDNA D-loop region as previously described (15). PCR was performed according to the protocol of the PCR Master Mix Kit (Promega, Madison, WI, USA) and purified prior to sequencing. Cycle sequencing was carried out using the Dye Terminator Cycle Sequencing Ready Reaction Kit (Applied Biosystems, Foster City, CA, USA) and the products were then separated on the ABI PRISM Genetic Analyzer 3100 (Applied Biosystems). Polymorphisms were confirmed by repeated analyses from the two strands. SNPs were identified directly from blood mito-chondria.

Statistical analysis

The age-at-onset curve of the HCC patients was calculated using the Kaplan-Meier method at each SNP site, and compared using the log-rank test. Multivariate survival analysis was performed using a Cox proportional hazards model. The statistical analyses were carried out using the SPSS 11.5 software package (SPSS Company, Chicago, IL, USA). A p-value of <0.05 was considered to indicate statistically significant differences.

Results

A total of 60 patients, including 49 HBV-associated and 11 alcohol-associated HCC patients, were enrolled in this study. The age-at-onset distribution of HCC patients is listed in Table I. Those analyzed included 6 patients aged <40 years, 12 patients aged 40–50, 30 patients aged 51–60 and 12 patients aged >60. None of these patients had received any adjuvant chemotherapy or radiation therapy following HCC resection. The age-at-onset and clinical characteristics of the HCC patients were analyzed using the Kaplan-Meier method and were compared using the log-rank test. Gender, portal vein thrombosis, child classification and tumor quantity were not associated with age-at-onset according to the results of the log-rank test. However, TNM classification and tumor size correlated with age-at-onset at statistically significant levels (Table II).

Table I.

Age-at-onset distribution in ESCC patients.

Table I.

Age-at-onset distribution in ESCC patients.

Age (years)No. of cases
MaleFemale
≤4042
41–50111
51–60282
>60102

[i] ESCC, esophageal squamous cell carcinoma.

Table II.

Clinical characteristics and their association with age-at-onset in HCC patients.

Table II.

Clinical characteristics and their association with age-at-onset in HCC patients.

CharacteristicsNo. of casesp-value
Gender0.370
  Male53
  Female7
TNM classification0.062
  I15
  II45
Portal vein thrombosis0.405
  Yes10
  No50
Child classification0.122
  A57
  B3
Tumor size (diameter)0.024
  <5 cm10
  ≥5 cm50
Tumor quantity0.271
  Single50
  Multiple10

[i] HCC, hepatocellular carcinoma.

Subsequently, the correlation between mtDNA genotype and age-at-onset was compared. We exluded the SNPs with a minor allele frequency of <5% and obtained 21 SNPs for further analysis. The HCC patients were divided into two groups on the basis of their genotype at each SNP site, and the age-at-onset curve was plotted using the Kaplan-Meier method for all HCC patients at these sites. A dramatic difference in age-at-onset appeared at sites 489 and 235 as shown by the log-rank test (Fig. 1). The 489C genotype, known as mtDNA haplogroup M, was significantly associated with a late age-at-onset of HCC when compared with halpogroup N (p=0.015) (Fig. 1). Another age-at-onset-associated SNP was identified at site 235 with frequency allele 235G and was linked with the late onset of HCC (p=0.009).

Using the Cox proportional hazards model, we performed multivariate analysis of HCC age-at-onset predictors including TNM classification, tumor size and the two SNPs. As shown in Table III, haplogroup M (489C) was identified as an independent predictive factor for age-at-onset of HCC at borderline significant levels [relative risk, 1.736; 95% confidence interval (CI), 0.967–3.115; p=0.065].

Table III.

Multivariate analysis of predictive factors associated with the age-at-onset of HCC.

Table III.

Multivariate analysis of predictive factors associated with the age-at-onset of HCC.

FactorsRelative risk95% CIp-value
Tumor size2.0230.831–4.9260.121
TNM classification1.1790.573–2.4270.654
235 (G/A)0.6110.254–1.4700.271
489 (C/T)1.7360.967–3.1150.065

[i] HCC, hepatocellular carcinoma; CI, confidence interval.

Discussion

Selected SNPs in the D-loop region have previously been examined for their ability to predict cancer risk and outcome in many types of cancer (2628). The present study has extended these analyses to determine the relationships between age-at-onset and germline SNPs in a continuous sequence of mtDNA between nucleotides 16190 and 583 in HCC patients. The SNPsm, 489C/T and 235G/A, were identified for their association with age-at-onset at statistically significant levels by the log-rank test. In an overall multivariate analysis, haplogroup M (489C) was identified as an independent predictive factor for age-at-onset of HCC at borderline significant levels. The results from the log-rank test revealed that disease advancement with a larger tumor size and a more serious clinical stage was more prevalent in younger patients. However, this was not shown in the multivariate analysis.

We have previously performed a number of studies on D-loop SNPs as predictive factors for digestive tract cancers (24,25,29). In the present study, for the first time, we suggest that, other than as predictors for cancer risk and outcome, SNPs in the D-loop are also predictors for age-at-onset in HCC patients. All non-African lineages belong to two founder clusters, namely haplogroups M and N; 489C, defined as haplogroup M, and 489T, defined as haplogroup N. According to the ‘out of Africa’ theory, these are both derived from the L3 mtDNA African lineage (30). The functional significance of the mtDNA haplo-groups and their association with human behaviors requires further study, although certain haplogroups have been identified as markers for special diseases (31,32). The results from this study require validation in the form of larger population sizes and laboratory-based functional studies.

The D-loop region of mtDNA is crucial for the regulation of mitochondrial genome replication and expression. SNPs in this region might affect mtDNA replication and lead to the alteration of the electron transport chain, which is responsible for the release of highly reactive oxygen species (ROS) and could contribute to nuclear genome damage as well as cancer initiation and promotion (3335). These two SNPs may alter the transcription of the mitochondrial genome, and this may subsequently enhance the production of ROS when the mitochondrial transcription is altered (36). These ROS-mediated mechanisms may thereby accelerate tumor development.

In conclusion, SNPs in the D-loop have been found to be biomarkers for the age-at-onset of HCC. The analysis of genetic polymorphisms in the D-loop might help to identify patient subgroups at high risk for an early onset, thereby helping to refine therapeutic decisions in HCC cancers.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (No. 30801384). The research was supported in part by the Natural Science Foundation of Hebei Province (No. C2008000958).

References

1. 

AI GomaaSA KhanMB ToledanoI WakedSD Taylor-Robinsonhepatocellular carcinoma: Epidemiology, risk factors and pathogenesisWorld J Gastroenterol1443004308200810.3748/wjg.14.430018666317

2. 

Z SunL MingX ZhuJ LuPrevetion and control of hepatitis B in ChinaJ Med Virol67447450200210.1002/jmv.1009412116043

3. 

J FerlayF BrayP PisaniDM ParkinGlobocan 2000: Cancer Incidence, Mortality and Prevalence Worldwide Version 1.0IARC PressLyon2001

4. 

S CaldwellSH ParkThe epidemiology of hepatocellular cancer: from the perspectives of public health problem to tumor biologyJ Gastroenterol4496101200910.1007/s00535-008-2258-619148801

5. 

A MakiH KonoM GuptaM AsakawaT SuzukiM MatsudaH FujiiI RusynPredictive power of biomarkers of oxidative stress and inflammation in patients with hepatitis C virus-associated hepatocellular carcinomaAnal Surg Oncol1411821190200710.1245/s10434-006-9049-117195915

6. 

S OkadaK ShimadaJ YamamotoT TakayamaT KosugeS YamasakiM SakamotoS HirohashiPredictive factors for postoperative recurrence of hepatocellular carcinomaGastroenterology1061618162419948194710

7. 

M MinagawaM MakuuchiT TakayamaN KokudoSelection criteria for repeat hepatectomy in patients with recurrent hepatocellular carcinomaAnn Surg238703710200310.1097/01.sla.0000094549.11754.e614578733

8. 

N WongW YeoWL WongNL WongKY ChanFK MoJ KohSL ChanAT ChanPB LaiTOP2A overexpression in hepatocellular carcinoma correlates with early age onset, shorter patients survival and chemoresistanceInt J Cancer124644652200910.1002/ijc.2396819003983

9. 

KB SchwarzOxidative stress during viral infection: a reviewFree Radical Biol Med21641649199610.1016/0891-5849(96)00131-18891667

10. 

A MansouriB FromentyA BersonMA RobinS GrimbertM BeaugrandS ErlingrD PessayreMultiple hepatic mitochondrial DNA deletions suggest premature oxidative aging in alcoholic patientsJ Hepatol2796102199710.1016/S0168-8278(97)80286-3

11. 

GS ShadelDA ClaytonMitochondrial DNA maintenance in vertebratesAnnu Rev Biochem66409435199710.1146/annurev.biochem.66.1.4099242913

12. 

S DiMauroEA SchonMitochondrial DNA mutations in human diseaseAm J Med Genet1061826200110.1002/ajmg.139211579421

13. 

MF BealMitochondia, free radicals, and neurodegenerationCurr Opin Neurobiol6661666199610.1016/S0959-4388(96)80100-08937831

14. 

RN LightowlersPF ChinneryDM TurnbullN HowellMammalian mitochondrial genetics: heredity, heteroplasmy and diseaseTrends Genet13450455199710.1016/S0168-9525(97)01266-39385842

15. 

DC WallaceMouse models for mitochondrial diseaseAm J Med Genet1067193200110.1002/ajmg.139311579427

16. 

MS FlissH UsadelOL CaballeroL WuMR ButaSM EleffJ JenD SidranskyFacile detection of mitochondrial DNA mutations in tumors and bodily fluidsScience28720172019200010.1126/science.287.5460.201710720328

17. 

S NomotoK YamashitaK KoshikawaA NakaoD SidranskyMitochondrial D-loop mutation as clonal markers in multicentric hepatocellular carcimona and plasmaClin Cancer Res8481487200211839667

18. 

E MamboX GaoY CohenZ GuoP TalalayD SidranskyElectrophile and oxidant damage of mitochondrial DNA leading to rapid evolution of homoplasmic mutationsProc Natl Acad Sci USA10018381843200310.1073/pnas.043791010012578990

19. 

H YoneyamaT HaraY KatoT YamoriET MatsuuraK KoikeNucleotide sequence variation is frequently in the mitochondrial DNA displacement loop region of individual human tumor cellsMol Cancer Res314202005

20. 

JP JakupciakS MaraghME MarkowitzAK GreenbergMO HoqueA MaitraPE BarkerPD WagnerWN RomS SrivastavaD SidranskyCD O’ConnellPerformance of mitochondrial DNA mutations detecting early stage cancerBMC Cancer8285200810.1186/1471-2407-8-28518834532

21. 

M NashikawaS NishiguchiS ShiomiA TamoriN KohT TakedaS KuboK HirohashiH KinoshitaE SatoM InoueSomatic mutation of mitochondrial DNA in cancerous and noncancerous liver tissue in individuals with hepatocellular carcinomaCancer Res6118431845200111280735

22. 

M Sanchez-CespedesP ParrellaS NomotoD CohenY XiaoM EstellerC JeronimoRC JordanT NicolWM KochM SchoenbergP MazzarelliVM FazioD SidranskyIdentification of a mononucleotide repeat as a major target for mitochondrial DNA alterations in human tumorsCancer Res6170157019200111585726

23. 

JW TaanmanThe mitochondrial genome: structure, transcription, translation and replicationBiochim Biophys Acta1410103123199910.1016/S0005-2728(98)00161-310076021

24. 

R ZhangF ZhangC WangS WangY-H ShiaoZ GuoIdentification of sequence polymorphism in the D-loop region of mitochondria DNA as a risk factor for hepatocellular carcinoma with distinct etiologyJ Exp Clin Cancer Res29130201010.1186/1756-9966-29-13020849651

25. 

C WangF ZhangH FanL PengR ZhangS LiuZ GuoSequence polymorphisms of mitochondrial D-loop and hepatocellular carcinoma outcomeBiochem Biophys Res Commun406493496201110.1016/j.bbrc.2011.02.08821345333

26. 

F NavagliaD BassoP FogarC SpertiE GrecoCF ZambonA StrangesA FaldaS PizziA ParentiS PedrazzoliM PlebaniMitochondrial DNA D-loop in pancreatic cancer: somatic mutations are epiphenomena while the germline 16519 T variant worsens metabolism and outcomeAm J Clin Pathol126593601200610.1309/GQFCCJMH5KHNVX7316938655

27. 

L WangWR BamletM de AndradeLA BoardmanJM CunninghamSN ThibodeauGM PetersenMitochondrial genetic polymorphisms and pancreatic cancer riskCancer Epidemiol Biomarkers Prev1614551459200710.1158/1055-9965.EPI-07-011917627010

28. 

L WangSK McDonnellSJ HebbringJM CunninghamJ St SauverJR CerhanG IsayaDJ SchaidSN ThibodeauPolymorphisms in mitochondrial genes and prostate cancer riskCancer Epidemiol Biomarkers Prev1735583566200810.1158/1055-9965.EPI-08-043419064571

29. 

R ZhangR WangF ZhangC WuH FanY liC WangZ GuoSingle nucleotide polymorphisms in the mitochondrial displacement loop and outcome of esophageal squamous cell carcinomaJ Exp Clin Cancer Res29155201010.1186/1756-9966-29-155

30. 

AM GonzálezJM LarrugaKK Abu-AmeroY ShiJ PestanoVM CabreraMitochondrial lineage M1 traces an early human backflow to AfricaBMC Genomics8223200717620140

31. 

V CarelliA AchilliML ValentinoC RengoO SeminoM PalaA OlivieriM MattiazziF PallottiF CarraraHaplogroup effects and recombination of mitochondrial DNA: novel clues from the analysis of Leber hereditary optic neuropathy pedigreesAm J Hum Genet78564574200610.1086/50123616532388

32. 

N UdarSR AtilanoM MemarzadehDS BoyerM ChwaS LuB MaguenJ LangbergP CoskunDC WallaceMitochondrial DNA haplogroups associated with age-related macular degenerationInvest Ophthalmol Vis Sci5019662974200910.1167/iovs.08-2646

33. 

B BandyAJ DavisionMitochondrial mutations may increase oxidative stress: implications for carcinogenesis and aging?Free Radic Biol Med8523539199010.1016/0891-5849(90)90152-92193852

34. 

JJ GilleH JoenjeCell culture models for oxidative stress: superoxide and hydrogen peroxide versus normobaric heperoxiaMutat Res275405414199210.1016/0921-8734(92)90043-O1383781

35. 

MK ShigenagaTM HagenBN AmesOxidative damage and mitochondrial decay in agingProc Natl Acad Sci USA911077110778199410.1073/pnas.91.23.107717971961

36. 

GA DementSC MaloneyR ReevesNuclear HMGA1 nonhistone chromatin proteins directly influence mitochondrial transcription, maintenance, and functionExp Cell Res3137787200710.1016/j.yexcr.2006.09.014

Related Articles

Journal Cover

March 2012
Volume 3 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Z, Yang H, Wang C and Liu S: Mitochondrial DNA haplogroup M is associated with late onset of hepatocellular carcinoma. Exp Ther Med 3: 499-502, 2012
APA
Guo, Z., Yang, H., Wang, C., & Liu, S. (2012). Mitochondrial DNA haplogroup M is associated with late onset of hepatocellular carcinoma. Experimental and Therapeutic Medicine, 3, 499-502. https://doi.org/10.3892/etm.2011.434
MLA
Guo, Z., Yang, H., Wang, C., Liu, S."Mitochondrial DNA haplogroup M is associated with late onset of hepatocellular carcinoma". Experimental and Therapeutic Medicine 3.3 (2012): 499-502.
Chicago
Guo, Z., Yang, H., Wang, C., Liu, S."Mitochondrial DNA haplogroup M is associated with late onset of hepatocellular carcinoma". Experimental and Therapeutic Medicine 3, no. 3 (2012): 499-502. https://doi.org/10.3892/etm.2011.434