research article

Association between Hepatitis C Virus Infection, P53 Phenotypes, and Gene Variants of Adenomatous Polyposis Coli in Hepatocellular Carcinomas

Leona N. Council1, Chandrakumar Shanmugam1,2, Esther A. Suswam1,3, Venkat R. Katkoori,1,4, Martine J. Heslin,5,3, Alex Hanna1, Nirag C. Jhala, 1,6, Sooryanarayana Varambally1,3 , UpenderManne*1,3

1Department of Pathology, University of Alabama at Birmingham, USA.

2Department ofSurgery, University of Alabama at Birmingham, USA.

3Comprehensive Cancer Center, University of Alabama at Birmingham, USA

Current Addresses: 2Department of Pathology, ESIC Medical College & Hospital, India.

4Department of Surgery, Michigan State University, USA.

6Department of Pathology and Laboratory Medicine, University of Pennsylvania, USA.

*Corresponding Author: UpenderManne, MS, PhD, Professor of Pathology, Room # 420A,Comprehensive Cancer Center, University of Alabama at Birmingham, 1802, 6th Avenue south – Physical, 1720 Second Avenue South – Mailing, Birmingham, AL 35294, USA. Tel: 205-934 4276; Fax: 205-975-6615; E-mail: upendermanne@uabmc.edu.

Received Date: 28 November, 2016; Accepted Date: 18 December, 2016; Published Date: 23 December, 2016

Citation: Council LN, Shanmugam C, Suswam EA, Katkoori VR, Manne U et al. (2016) Association between Hepatitis C Virus Infection, P53 Phenotypes, and Gene Variants of Adenomatous Polyposis Coli in Hepatocellular Carcinomas. J Dig Dis Hepatol 2016: JDDH-121.

Objective: To investigate the clinical value of p53 codon 72single nucleotide polymorphisms (SNPs) and variants of adenomatous polyposis coli (APC) in hepatocellular carcinomas (HCCs).

Methods: DNA and RNA from 51 HCCs and their matching, uninvolved liver tissues were analyzed for p53 mutations, and the methylation and expression of APC variants were determined. Proliferation of each HCC was assessed by Ki67 immunohistochemistry. The results were correlated with the demographic and clinicopathologic features and patient survival.

Results: Of 51 HCCs, 12% exhibited missense p53 mutations. SNP analysis of p53 codon 72 demonstrated the highest prevalence of the Arg/Arg (56%) phenotype, followed by Arg/Pro (33%) and Pro/Pro (11%). Four of five cases with the Pro/Pro phenotype were African Americans (AAs). All five cases with the Pro/Pro phenotype had hepatitis C virus (HCV) infections, a high Ki67 index, and lower median survival (15.5 months) compared to those with Arg/Arg or Arg/Pro phenotypes (32 months). The overall frequency of APC methylation was 31%, which was found predominantly in Caucasians. There was lower mRNA expression of APC variants-2 and -3 in both HCCs and corresponding adjacent, uninvolved liver tissues as compared to APC variant-1. The expression of APC variant-3, but not variants-1 and -2, was lower in HCCs relative to uninvolved tissues. Expression of all APC variants was lower in HCCs with APC methylation relative to HCCs without APC methylation, and low expression of APC variant-2 was associated with the Pro/Pro phenotype.

Conclusions: These findings suggest that, for AA patients with HCCs,the p53 Pro/Pro phenotype and low expression of APC variant-2 are associated withaggressive tumor behavior, HCV infection, and poor clinical outcome.

Keywords: Hepatocellular carcinoma; APC; p53 codon 72 polymorphism; HCV; Survival

Background

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide  In the United States, there were 26,190 new cases and 19,590 deaths due to HCCs in 2011 [6]. Both the incidence and mortality of this disease are increasing [7, 8] primarily due to hepatitis C virus (HCV) infections  Worldwide, hepatitis B virus (HBV) and HCV infections, cigarette smoking, and/or heavy alcohol consumption are factors contributing to HCCs [10]. Although surgery is useful in some cases, cirrhosis in the adjacent, uninvolved liver tissue predisposes patients to metachronous HCCs. Thus, there is a need for biomarkers that can be used in assessment of the prognosis and in treatment decisions.

The pathogenesis of HCCs involves genetic alterations; irrespective of etiology, there is loss of p53 function The p53 mutational spectra vary with geographic regions, based on etiological differences and host susceptibility. People in sub-Saharan Africa and China, who have endemic exposure to aflatoxin B1 (AFB1) have a higher mutation frequency at codon 249 in p53 [12]. In the USA and Europe, p53 mutations with no specific hotspot occur in HCCs, either along with or after loss of heterozygosity (LOH) of p53 [13]. For HCCs, p53 mutations correlate with disease recurrence and poor prognosis [14]. In addition, the HBx gene of HBV is integrated into the host genome and binds to p53 to suppress its DNA binding and transcription as well as p53-mediated apoptosis [15]. Although the HCV genome does not integrate into the host genome, it suppresses the transcriptional activity of the p53 promoter [16]. The codon 72 polymorphism of p53 results in Pro/Pro and Arg/Arg phenotypes, with the Pro/Pro phenotype having less apoptosis-inducing potential and higher cancer cell proliferation p53 codon 72 polymorphisms are associated with risk of various cancers, including those of the liver [19, 20]. The p53 codon 72 Pro/Pro phenotype is associated with increased risk of tumor progression and poor patient survival for non–small cell lung cancers [21]and colorectal cancers (CRCs), particularly for (AAs) 

The adenomatous polyposis coli (APC) protein functions as a tumor suppressor by forming a tetramer with β-catenin, conductin (a negative regulator of Wnt pathway), and serine-threonine glycogen synthase kinase-3β, which enhances β-catenin degradation, thereby preventing signal transduction to downstream transcription factors  This function is altered due to mutations [24]and/or LOH [25]in the APC gene, and/or to APC promoter hypermethylation  Unlike for tumors of other anatomic sites, mutations of the APC gene have not been identified in HCCs; however, APC promoter hypermethylation is frequent . The present investigation focused on assessing the clinical value of the p53 codon 72 polymorphism, APC promoter hypermethylation, and expression of APC variants together in HCCs.

Materials and Methods Patients and tissues  

This study was approved by the Institutional Review Board of the University of Alabama at Birmingham (UAB). Medical records, including surgical pathology reports, were reviewed by two gastrointestinal pathologists (LNC & CS). From 137 HCC patients who had undergone transplantation or resection as a curative procedure between 1991 and 2009, formalin-fixed, paraffin embedded (FFPE) tissue samples were obtained for 51 cases. None of these patients had received pre-surgical chemo- or radiation therapy. Diagnosis for chronic HCV infection was assessed based on the anti-HCV antibody status of the patients. The pathologists also reviewed hematoxylin and eosin-stained slides for pathologic features. HCCs were gradedas well, moderate, or poorly differentiated [28],and staged by the criteria of the American Joint Commission on Cancer [29].

This patient population consisted of 15 AAs and 34 non-Hispanic Caucasians (CAs); for 2 patients, racial/ethnic information was missing. All patients were followed by the UAB tumor registry until their death or the date of the last documented contact within the study time frame. The registry also ascertained this information directly from patients or living relatives and/or from physician charts and validated it against State Death Lists. The registry updated information every 6 months; follow-up ended in December 2011.

p53 gene mutational analysis 30]. Of these, 48 DNA samples were analyzable for p53 mutations (exon 4 through 9) by the polymerase chain reaction (PCR) and direct sequencing using exon-specific primers, as described in our previous report  In brief, the PCR products were purified by electrophoresis on 3% agarose gels. The purified products were sequenced with an ABI 3100 sequencer. Compilation and sequence analyses were accomplished with LASERGENE (DNA STAR Inc., Madison, WI) software, which allows direct analysis of sequencing electrophoretograms for detection of duplex sequence signals at each position to identify mutations/polymorphisms. Nucleotide changes in each exon sequence were confirmed by sequencing the opposite strand.

Methylation status of APC gene 

DNA methylation status in the CpG island of the APC gene promoter was determined by chemical modification of the unmethylated, but not the methylated, cytosines to uracil, and subsequent PCR using primers specific for either the methylated or the modified unmethylatedDNA [31]. Primer sequences of APC were, for the unmethylated reaction, 5′-gtgttttattgtggagtgtgggtt-3′ (forward primer) and 5′-ccaatcaacaaactcccaacaaa-3′ (reverse primer) and, for the methylated reaction, 5′-tattgcggagtgcgggtg-3′ (forward primer) and 5′-tcgacgaactcccgacga-3′ (reverse primer). Briefly, 1 μg of DNA was modified and purified with an imprint DNA modification kit (Sigma-Aldrich, Saint Louis, MO). Controls without DNA were performed for each set of primers. Portions of each PCR reaction (10 µL) were loaded onto 3% agarose gels, stained with ethidium bromide, and visualized under UV illumination.

APC gene expression analysis 

Total RNA from 51 HCCs and matching uninvolved tissues was extracted using RNeasy Kits (Qiagen Life Sciences, Valencia, CA), and 1 µg of RNA was reverse transcribed. For each HCC and matching uninvolved tissue, the expression levels of three previously identified transcript forms of the APC gene (variant-1, NM_001127511.2; variant-2, NM_001127510.2; and variant-3, NM_000038.5) ]were determined by quantitative real-time PCR (qRT-PCR) with transcript-specific primers. Briefly, PCR was performed with SYBR green reagent supermix (Bio-Rad laboratories, Hercules, CA) in a final volume of 25 µL consisting of 0.5 µL of each primer (5 pmoles), 12.5 µL of 2x supermix containing the reaction buffer, Fast-start Tag DNA double strand-specified SYBR green I dye, 7.0 µL of nuclease-free water, and 5 µL of the cDNA template. Incubation was at 95°C for an initial denaturation, followed by 45 cycles of 15-sec denaturations at 95°C, and annealing and extension at 60°C for 30 sec. The PCR reactions, performed in sets of four, were accomplished with an i-Cycler real-time PCR system (Bio-Rad). To exclude non-specific amplification, PCR products were subjected to melting curve analysis. For each patient, means of APC gene transcripts and β-actin gene copy numbers were calculated, and ratios of these means (APC/β-actin) were generated.

Immunohistochemistry

Sections (5-µm) representative of uninvolved and tumor tissue were cut from FFPE blocks and mounted on Superfrost/Plus slides (Fisher Scientific; Pittsburgh, PA). In brief, sections were melted at 60°C for 1.5 hr and incubated overnight at 37°C prior to staining. Sections were then deparaffinized in xylene, rehydrated in graded alcohols, and transferred to a Tris-buffer bath (0.05M Tris base, 0.15 M NaCl, and 0.01% Triton X-100, pH 7.6). Heat-induced antigen retrieval was performed with a pressure cooker at 15 psi for 10 min with EDTA buffer (0.01 M, pH 9). Each section was treated with 3% H2O2 for 5 min to quench endogenous peroxidase and incubated with 3% goat serum at room temperature for 1 hr to reduce nonspecific immunostaining. Tissue sections were incubated with antihuman Ki67 primary monoclonal antibody (clone SP6, dilution 1:1200; Lab Vision, Fremont, CA) raised in rabbits. Sections to which the primary antibody was not applied were utilized as negative controls. Secondary detection was accomplished with a horseradish peroxidase-labeled anti-rabbit antibody for 40 min (Jackson Immuno Research, West Grove, PA). Diaminobenzidine tetrachloride supersensitive substrate kits (ScyTek, Logan, UT) were used to visualize the antibody–antigen complex. Each section was counterstained with hematoxylin, dehydrated with graded alcohols, and washed with xylene before mounting with coverslips. Slides were independently examined and scored by two pathologists (LNC & CS). For discrepancies in individual scores, the pathologists re-evaluated these together to reach a consensus before combining the final individual scores. As described in previous studies , only tumors with distinct nuclear immunostaining in ≥10% of the cells were considered as positive.

Statistical analyses

Chi-square analyses were used to correlate the alterations of p53 and APC genes with clinical and pathological variables. P values of < 0.05 were considered statistically significant.

Results 

Study cohort characteristics  

The median age of the study cohort was 57 (range, 13-81) years; 35 were male, and 16 were female (Table 1). Most HCCs were associated with HCV infection (37 of 51, 73%) and alcohol use (32 of 51, 63%). The median survival of the cohort was 30 months (range, 2-146 months).

p53 mutations and codon 72 polymorphism  

The overall incidence of missense point mutations of p53 (Table 2) was low (6 of 48, 12%). Missense point mutations, however, were common in CAs (4 of 6), and almost all of these patients had HCV infection (Table 2). CAs with point mutations had shorter median survival (22.5 months) relative to those with wild-type p53 (34 months) (data not shown).

The sequence traces representing the Arg/Arg, Arg/Pro, and Pro/Pro phenotypes of p53 codon 72 in HCCs are shown in Figure 1. In these analyses, the Arg/Argphenotype had the highest prevalence (27 of 48, 56%) followed by Arg/Pro (16 of 48, 33%) and Pro/Pro (5 of 48, 11%) (Table 3). Four of five patients with the Pro/Pro phenotype were AAs (χ2 p = 0.028). All AA patients with the Pro/Pro phenotype had high Ki67 indices (Figure 2). Poorly differentiated tumors were most prevalent in Pro/Pro phenotype (2 of 5, 40%) (χ2 p = 0.016) and less prevalent in patients with Arg/Arg (1 of 27, 4%) or Arg/Pro (1 of 16; 6%) phenotypes (Table 3). All patients with the Pro/Pro phenotype had HCV infections (χ2 p = 0.048) (Table 3). Furthermore, AAs with the Pro/Pro phenotype had lower median survival (15.5 months) relative to those with Arg/Arg or Arg/Pro (32 months) phenotype (data not shown).

< >APC gene methylation and mRNA expressionDiscussion34], but similar to a low p53 mutation frequency in Caucasian patients from Italy (13%) In related studies, a high incidence of missense mutations at p53 codon 249 was found in geographic regions with high AFB1 exposure, unlike the United States, where AFB1 exposure is rare [12]. Our study found p53 codon 249 mutation in only one patient. This geographical discrepancy in p53 mutations may be due to variations in etiological factors and/or  individual susceptibility based on race/ethnicity. For many cancers, including HCCs, p53 mutations are associated with poor prognoses Our results demonstrated a shorter median survival for patients with p53 mutations, particularly CAs; however, due to the low frequency of p53 mutations in tumors of AA patients, a prognostic value for these mutations could not be demonstrated.

The codon 72 SNP of p53 results in Pro/Pro and Arg/Argphenotypes. These phenotypes are distinct in that the Arg/Argphenotype, owing to its localization in mitochondria, induces apoptosis 15-fold more than the Pro/Pro phenotype . These phenotypes also differ in their biological and biochemical activities In our study, the Arg/Argphenotype had the highest prevalence (56%) in HCCs, followed by Arg/Pro (33%) and Pro/Pro (11%). This is in concordance with other reports for HCCs in which the prevalence of the Arg/Arg and Pro/Pro phenotypes was 54.2% and 13.5%, respectively [39]. In our study, most patients exhibiting the Pro/Pro phenotype were AAs, whose tumors had higher proliferation rates and who had a shorter median survival. Similarly, for CRCs, thePro/Pro phenotype, relative to the Arg/Argphenotype, was frequent in AAs and was associated with aggressive tumors and a poor prognosis  Thus, the Pro/Pro phenotype at codon 72 of p53 is a race-specific, molecular prognostic marker for AA patients with CRCs and HCCs. These and other studies aid in identifying unique germline alleles/allelic combinations that modify cancer risk and/or cancer progression. Furthermore, the differences between racial/ethnic groups illustrate the effect of different lifestyles and/or exposures to environmental factors 

Compared to HBV, HCV infections induce inflammation and can lead to cirrhosis and to HCC HCV infections suppress the transcriptional activity of the p53 promoter [16]. Gene expression studies of HCV-related cirrhosis reveal up-regulation of pro-inflammatory, pro-apoptotic, and pro-proliferative genes Furthermore, induction of the HCV core protein in primary hepatocytes during senescence, immortalization, and anchorage-independent growth involves pathways that regulate cellular growth, immune response, oxidative stress, and apoptosis In our study, all HCC patients exhibiting the Pro/Pro phenotype had HCV infections. Furthermore, the Pro/Pro phenotype was associated with increased risk of HCC in HCV-negative cases [19, 44]. Together, these results suggest that the risk of HCC from the Pro/Pro phenotype is influenced by a chronic immune response against HCV, which may either conceal  or exacerbate the effect of the Pro/Pro phenotype , especially in AA patients with chronic liver disease, for which HCV is the most common cause [47].

The APC gene is involved in transduction of Wnt-signaling [23, 52, 53]. Unlike in CRCs, APC mutations are rare in other gastrointestinal malignancies, including HCCs [12], hence, we did not perform APC mutational analyses. However, APC promoter hypermethylation, a common phenomenon in HCCs, is reported in 53 to 100% of HCCs and in 27% of cases of liver cirrhosis In our study, 31% of HCCs showed APChypermethylation, with CA patients having a higher frequency relative to AAs. The disparity in frequency of APC methylation between CAs and AAs, as shown for CRCs, may be due to differences in dietary factors, such as alcohol consumption 

Lower expression of tumor suppressor genes, either due to the influence of allelic loss or epigenetic mechanisms, is common in malignancies, including HCCs  The present study is first to demonstrate lower mRNA expression of APC variants-2 and -3 in both HCCs and corresponding adjacent uninvolved liver tissues as compared to variant-1. In addition, there were lower levels of APC variants-2 and -3 in HCCs with APC promoter methylation as compared to HCCs without methylation. This suggests that APC methylation alters the expression of APC variants -2 and -3. Since splice variants of untranslated leader exons are associated with alternative promoter usage, resulting in differential gene expression with respect to cell or tissue type, efficient translation of the mRNA depends on differing leader exons and the methylation status of the involved gene [51]. The usage of alternative promoters in the regulation of APC variant expression may explain the observed differential expression of APC variants. These results demonstrate that low expression of APC variant-2 is associated with the Pro/Pro phenotype. Future studies of HCCs should identify the location of alternative promoters or genetic and epigenetic events involved in the control of expressionof these APC variants and their relationship with the Pro/Pro phenotype of p53. In the current study, samples were collected from transplants or resections; hence, there is a possibility to have more favorable outcomes than those observed in the general population. However, the present results form a basis for large retrospective population-based studies or prospective studies to validate these findings.

Conclusions Competing Interests:None Financial disclosures:None Author contributions:Leona N. Council, ChandrakumarShanmugam, and Venkat R. Katkoori contributed equally to this work; Leona N. Council, ChandrakumarShanmugam, Venkat R. Katkoori, Martine J. Heslin, Nirag C. Jhala and UpenderManne designed the research; Leona N. Council, ChandrakumarShanmugam, and Venkat R. Katkoori performed the research; Leona N. Council, ChandrakumarShanmugam, Venkat R. Katkoori, Alex Hanna, and UpenderManne analyzed the data; and Leona N. Council, ChandrakumarShanmugam, Venkat R. Katkoori, Esther A. Suswam, and UpenderManne wrote the manuscript.

Acknowledgements

We thank Dr. Donald L. Hill, Division of Preventive Medicine, University of Alabama at Birmingham, AL, for his critical review of this manuscript.

Funding sources: This study was supported jointly by a Charles Barkley Award through the UAB Minority Health Disparities Research Center and a pilot grant from the UAB Center for Clinical and Translational Science (5UL1 RR025777) to Dr. Leona N. Council, and in part by grants from the NIH (3U54-CA118948, R01-CA98932, R03-CA139629) to Dr. Manne.

Figure 1: Genotypic analysis for codon 72 of the p53 gene.

Direct sequencing analysis of the p53 gene at codon 72 for HCCs and uninvolved tissues demonstrating a nucleotide change (G>C). (A) homozygous for G/G (Arg/Arg phenotype), (B) heterozygous for G/C (Arg/Pro phenotype), (C) homozygous for C/C (Pro/Pro phenotype).

 

Figure 2: Ki-67 immunohistochemistry of HCCs based on p53 codon 72 phenotypes.

Nuclear positivity (thick arrows) for Ki67 demonstrating low and high proliferative index in HCCs with Arg/Arg (A, 200 μm) and Pro/Pro (B, 200 μm) phenotypes. Note occasional positivity (thin arrows) in the adjacent uninvolved tissues (B).

 

Figure 3: Expression of APC variants and its association with APC methylation in HCCs and their adjacent, uninvolved liver.

(A) mRNA expression of APC variants in HCCs and uninvolved tissues. Variant 3 (V3) demonstrated lower levels of expression in HCCs (T) and their corresponding, uninvolved tissues (N) relative to variants 1 and 2 (V1 & V2). (B) mRNA expression of APC variants in HCCs based on APC promoter methylation status. The APC variants 2 and 3 (V2 & V3) demonstrated lower expression in HCCs exhibiting APC methylation (M) as compared to HCCs without APC methylation (UM).

 

 

 

 

Variable

 

Number of patients (%)

 

Age group (years)

 

< 50

7 (14)

≥ 50

44 (86)

Gender

 

Female

16 (31)

Male

35 (69)

Ethnicitya

 

African Americans (AAs)

15 (29)

Caucasians (CAs)

34 (67)

HCV Status

 

Positive

37 (73)

Negative

14 (27)

Tumor stagea

 

I

35 (69)

II

11 (21)

III

3 (6)

IV

1 (2)

Tumor differentiation

 

Well

21 (41)

Moderate

26 (52)

Poor

4 (8)

Alcohol usea

 

No

17 (33)

Yes

32 (63)

aInformation is missing on race/ethnicity and alcohol usage for two cases, and tumor staging for one case.

Table 1: Clinicopathological features of the HCC study cohort (N= 51)

 

 

Case no.

 

Mutation at codon

 

Race/ ethnicity

 

Nucleotide change

 

Amino Acid change

 

Mutation type

 

Codon 72 phenotype

 

HCV status

5

308

CA

GCA>TCA

Ala>Ser

Missense

Arg/Arg

HCV

19

291

CA

CGC>TGC

Arg>Cys

Missense

Arg/Pro

None

24

235

CA

AAC>TAC

Asn>Tyr

Missense

Arg/Arg

HCV

25

249

NA

AGG>AGT

Arg>Ser

Missense

Arg/Arg

None

26

111

CA

CTG>CTA

Leu>Leu

Silent

Arg/Arg

HCV

27

246

CA

ATG>GTG

Met>Val

Missense

Arg/Arg

HCV

28

121

CA

TCT>TCA

Ser>ser

Silent

Arg/Arg

HCV

32

219

AA

CCC>GCC

Pro>Ala

Missense

Pro/Pro

HCV

45

88

CA

GCC>GCT

Ala>Ala

Silent

Arg/Arg

HCV

CA- Caucasian; AA- African-American; HCV- Hepatitis C Virus; NA- Information not available

Table 2: Details of p53 alterations and HCV infection

 

 

Variable

 

Number of patients (N=48)

 

χ2 P-value

 

Arg/Arg

Arg/Pro

Pro/Pro

 
 

27 (56%)

16 (33%)

5 (11%)

 

Age group (years)

       

< 50

4 (15)

3 (19)

0 (0)

0.583

≥ 50

23 (85)

13 (81)

5 (100)

Gender

       

Female

8 (30)

6 (38)

2 (40)

0.822

Male

19 (70)

10 (62)

3 (60)

Ethnicitya

       

African-Americans

5 (19)

5 (31)

4 (80)

0.028

Caucasians

20 (74)

11 (69)

1 (20)

HCV status

       

Positive

21 (78)

8 (50)

5 (100)

0.048

Negative

6 (22)

8 (50)

0 (0)

Tumor stagea

       

I

17 (81)

13 (65)

2 (40)

0.295

II

7 (19)

1 (24)

3 (60)

III

2 (0)

1 (9)

0 (0)

 

IV

1 (0)

0 (0)

0 (0)

 

Tumor differentiation

       

Well

13 (48)

4 (25)

3 (60)

0.016

Moderate

13 (48)

11 (69)

0 (0)

Poor

1 (4)

1 (6)

2 (40)

 

Alcohol usea

       

No

9 (33)

6 (38)

1 (20)

0.718

Yes

17 (63)

9 (56)

4 (80)

APC variant expressiona

       

Variant-1

       

High

11 (41)

9 (56)

4 (80)

0.226

Low

16 (59)

7 (44)

1 (20)

Variant-2

       

High

12 (44)

12 (75)

1 (20)

0.048

Low

15 (56)

4 (25)

4 (80)

Variant-3

       

High

10 (37)

8 (50)

3 (60)

0.525

Low

17 (63)

8 (50)

2 (40)

Mortality

       

Alive

24(89)

14(88)

4(80)

0.915

Dead

3(11)

2(12)

1(20)

aInformation is missing on race/ethnicity and alcohol usage for two cases and tumor staging for one case.

bhigh (median value of β-actin/APC mRNA ratio greater than corresponding normal) expressors

Table 3: Association between p53 polymorphisms and clinicopathological characteristics

 

 

Variable

 

Number of patients (N=51)

 

χ2 P-value

 

Methylated

Unmethylated

 
 

APC

APC

 
 

16 (31%)

35 (69%)

 

Age group (years)

     

< 50

3 (19)

4 (11)

0.48

≥ 50

13 (81)

31(89)

Gender

     

Female

5 (31)

11 (31)

1

Male

11 (69)

24 (69)

Ethnicitya

     

African-Americans

1 (6)

14 (40)

0.024

Caucasians

13 (81)

21 (60)

HCV status

     

Positive

13 (81)

24 (69)

0.346

Negative

3 (19)

11 (31)

Tumor stagea

     

I

13 (81)

22 (65)

0.489

II

3 (19)

8 (24)

III

0 (0)

3 (9)

IV

0 (0)

1 (2)

Tumor differentiation

     

Well

8 (50)

13 (37)

0.321

Moderate

8 (50)

18 (51)

Poor

0 (0)

4 (12)

Alcohol usea

     

No

3 (19)

14 (40)

0.151

Yes

12 (75)

20 (57)

Mortality

     

Alive

14(88)

28(88)

0.643

Dead

2(12)

4(12)

aInformation is missing on race/ethnicity and alcohol usage for two cases, and tumor staging of one case.

Table 4: Association between APC methylation status and clinicopathological characteristics

 

  1. El-Serag HB (2012) Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 142: 1264-1273.
  2. Ferlay J, SoerjomataramI, Dikshit R, Eser S, Mathers C (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer136:E359–386.
  3. Mittal S, El-Sareg HB (2013)Epidemiology of HCC: Consider the Population. J ClinGastroenterol 47:S2–S6.
  4. Takeda H, Takai A, Inuzuka T, Marusawa H (2016) Genetic basis of hepatitis virus-associated hepatocellular carcinoma: linkage between infection, inflammation, and tumorigenesis. J Gastroenterol Oct 6, (Epub ahead of print):1-13.
  5. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J et al. (2015) Global cancer statistics, 2012. CA Cancer J Clin 65: 87-108.
  6. Siegel R, Ward E, Brawley O, Jemal A (2011) Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61:212-236.
  7. Altekruse SF, Henley SJ, Cucinelli JE, McGlynn KA (2014) Changing hepatocellular carcinoma incidence and liver cancer mortality rates in the United States. Am J Gastroenterol 109:542-553.
  8. Petrick JL, Kelly SP, Altekruse SF, McGlynn KA, Rosenberg PS (2016) Future of Hepatocellular Carcinoma Incidence in the United States Forecast Through 2030. J ClinOncol 34: 1787-1794.
  9. El-Serag HB and Mason AC (1999) Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med 340: 745-750.
  10. Yu MC and Yuan JM (2004) Environmental factors and risk for hepatocellular carcinoma. Gastroenterology 127: S72-78.
  11. Edamoto Y, Hara A, Biernat W, Terracciano L, Cathomas G et al. (2003) Alterations of RB1, p53 and Wnt pathways in hepatocellular carcinomas associated with hepatitis C, hepatitis B and alcoholic liver cirrhosis. Int J Cancer 106:334-341.
  12. Teufel A, Staib F, Kanzler S, Weinmann A, Schulze-Bergkamen H et al. (2007) Genetics of hepatocellular carcinoma. World J Gastroenterol 13: 2271-2282.
  13. Oda T, Tsuda H, Scarpa A, Sakamoto M, Hirohashi S (1992) p53 gene mutation spectrum in hepatocellular carcinoma. Cancer Res 52: 6358-6364.
  14. Qin LX and Tang ZY (2002) The prognostic molecular markers in hepatocellular carcinoma. World J Gastroenterol 8: 385-392.
  15. Lee SG and Rho HM (2000) Transcriptional repression of the human p53 gene by hepatitis B viral X protein. Oncogene 19:468-471.
  16. Ray RB, Steele R, Meyer K, Ray R (1997) Transcriptional repression of p53 promoter by hepatitis C virus core protein. J BiolChem 272:10983-10986.
  17. Dumont P, Leu JI, Della Pietra AC, George DL, Murphy M (2003) The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nat Genet 33:357-365.
  18. Pim D and Banks L (2004) p53 polymorphic variants at codon 72 exert different effects on cell cycle progression. Int J Cancer 108: 196-199.
  19. Ezzikouri S, El Feydi AE, Chafik A, Benazzouz M, El Kihal L et al. (2007) The Pro variant of the p53 codon 72 polymorphism is associated with hepatocellular carcinoma in Moroccan population. Hepatol Res 37: 748-754.
  20. Sumbul AT, Akkiz H, Bayram S, Bekar A, Akgollu E et al. (2012) p53 codon 72 polymorphism is associated with susceptibility to hepatocellular carcinoma in the Turkish population: a case-control study. MolBiol Rep 39:1639–1647.
  21. Wang YC, Lee HS, Chen SK, Chang YY, Chen CY (1999) Prognostic significance of p53 codon 72 polymorphism in lung carcinomas. Eur J Cancer 35: 226-230.
  22. Katkoori VR, Jia X, Shanmugam C, Wan W, Meleth S et al. (2009) Prognostic significance of p53 codon 72 polymorphism differs with race in colorectal adenocarcinoma. Clin Cancer Res 15:2406-2416.
  23. Behrens J, Jerchow BA, Würtele M, Grimm J, Asbrand C et al. (1998) Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta. Science 280: 596-599.
  24. Michils G, Tejpar S, Fryns JP, Legius E, Van Cutsem E (2002) Pathogenic mutations and rare variants of the APC gene identified in 75 Belgian patients with familial adenomatous polyposis by fluorescent enzymatic mutation detection (EMD). Eur J Hum Genet 10:505-510.
  25. Hiltunen MO, Alhonen L, Koistinaho J, Myohanen S, Paakkonen M et al. (1997) Hypermethylation of the APC (adenomatous polyposis coli) gene promoter region in human colorectal carcinoma. Int J Cancer 70:644-648.
  26. EstellerM, Sparks A, Toyota M, Sanchez-Cespedes M, Capella G et al. (2000) Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res 60:4366-4371.
  27. Csepregi A, Rocken C, Hoffmann J, Gu P, Saliger S et al. (2008) APC promoter methylation and protein expression in hepatocellular carcinoma. J Cancer Res ClinOncol134:579-589.
  28. Bosman FT, Carneiro F, Hruban RH, Theise ND (2010) WHO Classification of Tumours of the Digestive System (IARC WHO Classification of Tumours) 4th ed: IARC.
  29. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL et al. (2010) American Joint Committee on Cancer, American Cancer Society: AJCC Cancer Staging Manual, 7th ed. New York, NY: Springer-Verlag.
  30. Fredricks DN and Relman DA (1999) Paraffin removal from tissue sections for digestion and PCR analysis. Biotechniques 26: 198-200.
  31. Lambertz S and Ballhausen WG (1993) Identification of an alternative 5' untranslated region of the adenomatous polyposis coli gene. Hum Genet 90: 650-652.
  32. Jin Z, Tamura G, Tsuchiya T, Sakata K, Kashiwaba M et al. (2001) Adenomatous polyposis coli (APC) gene promoter hypermethylation in primary breast cancers. Br J Cancer 85: 69-73.
  33. Tan PH, Bay BH, Yip G, Selvarajan S, Tan P (2005) Immunohistochemical detection of Ki67 in breast cancer correlates with transcriptional regulation of genes related to apoptosis and cell death. Mod Pathol 18:374-381.
  34. Hsu H, Peng S, Lai P, Chu J, Lee P (1994) Mutations of p53 gene in hepatocellular-carcinoma (hcc) correlate with tumor progression and patient prognosis - a study of 138 patients with unifocalhcc. Int J Oncol 4:1341-1347.
  35. Pontisso P, Belluco C, Bertorelle R, De Moliner L, Chieco-Bianchi L (1998) Hepatitis C virus infection associated with human hepatocellular carcinoma: lack of correlation with p53 abnormalities in Caucasian patients. Cancer83:1489-1494.
  36. Dou J, Liu P, Zhang X (2005) Cellular response to gene expression profiles of different hepatitis C virus core proteins in the Huh-7 cell line with microarray analysis. J NanosciNanotechnol5:1230-1235.
  37. Fukutomi T, Zhou Y, Kawai S, Eguchi H, Wands JR (2005) Hepatitis C virus core protein stimulates hepatocyte growth: correlation with upregulation of wnt-1 expression. Hepatology 41:1096-1105.
  38. Marin MC, Jost CA, Brooks LA, Irwin MS, O'Nions J et al. (2000) A common polymorphism acts as an intragenic modifier of mutant p53 behaviour. Nat Genet 25:47-54.
  39. Ezzikouri S, El Feydi AE, Chafik A, Benazzouz M, El Kihal L (2007) The Pro variant of the p53 codon 72 polymorphism is associated with hepatocellular carcinoma in Moroccan population. Hepatol Res 37:748-754.
  40. Slattery ML, Schaffer D, Edwards SL, Ma KN, Potter JD (1997) Are dietary factors involved in DNA methylation associated with colon cancer? Nutr Cancer 28:52-62.
  41. Ikeda K, Saitoh S, Koida I, Arase Y, Tsubota A et al. (1993) A multivariate analysis of risk factors for hepatocellular carcinogenesis: a prospective observation of 795 patients with viral and alcoholic cirrhosis. Hepatology 18:47-53.
  42. Tomoda T, Nouso K, Sakai A, Ouchida M, Kobayashi S et al. (2012) Genetic risk of hepatocellular carcinoma in patients with hepatitis C virus: a case control study. J GastroenterolHepatol27(4):797–804.
  43. Shackel NA, McGuinness PH, Abbott CA, Gorrell MD, McCaughan GW (2002) Insights into the pathobiology of hepatitis C virus-associated cirrhosis: analysis of intrahepatic differential gene expression. Am J Pathol 160:641-654.
  44. Hu S, Zhao L, Yang J, Hu M (2014) The association between polymorphism of P53 Codon72 Arg/Pro and hepatocellular carcinoma susceptibility: evidence from a meta-analysis of 15 studies with 3,704 cases. TumourBiol 35:3647–3656.
  45. Yano Y, Yamashita F, Kuwaki K, Fukumori K, Kato O (2006) Clinical features of hepatitis C virus-related hepatocellular carcinoma and their association with alpha-fetoprotein and protein induced by vitamin K absence or antagonist-II. Liver Int 26:789-795.
  46. Zhu N, Khoshnan A, Schneider R, Matsumoto M, Dennert G et al. (1998) Hepatitis C virus core protein binds to the cytoplasmic domain of tumor necrosis factor (TNF) receptor 1 and enhances TNF-induced apoptosis. J Virol 72:3691-3697.
  47. Chen SL and Morgan TR (2006) The Natural History of Hepatitis C virus (HCV) infection. Int J Med Sci 3: 47-52.
  48. Lee S, Lee HJ, Kim JH, Lee HS, Jang JJ (2003) Aberrant CpG island hypermethylation along multistep hepatocarcinogenesis. Am J Pathol 163:1371-1378.
  49. van Engeland M, Weijenberg MP, Roemen GM, Brink M, de Bruine AP et al. (2003) Effects of dietary folate and alcohol intake on promoter methylation in sporadic colorectal cancer: the Netherlands cohort study on diet and cancer. Cancer Res63:3133-3137.
  50. Virmani AK, Rathi A, Sathyanarayana UG, Padar A, Huang CX et al. (2001) Aberrant methylation of the adenomatous polyposis coli (APC) gene promoter 1A in breast and lung carcinomas. Clin Cancer Res 7: 1998-2004.
  51. Ayoubi TA and Van De Ven WJ (1996) Regulation of gene expression by alternative promoters. FASEB J 10: 453-460.
  52. Ding SL, Yang ZW, Wang J, Zhang XL, Chen XM et al. (2015) Integrative analysis of aberrant Wnt signaling in hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol 21:6317-28.
  53. Benchabane H and Ahmed Y (2009) The adenomatous polyposis coli tumor suppressor and Wnt signaling in the regulation of apoptosis. AdvExp Med Biol 656: 75-84.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Digestive Diseases and Hepatology

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131