Mutational and promoter hypermethylation status of FHIT gene in breast cancer patients of Kashmir

8
Please cite this article in press as: N. Syeed, et al., Mutational and promoter hypermethylation status of FHIT gene in breast cancer patients of Kashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j.mrfmmm.2010.11.001 ARTICLE IN PRESS G Model MUT 10993 1–8 Mutation Research xxx (2010) xxx–xxx 1 Contents lists available at ScienceDirect Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis journal homepage: www.elsevier.com/locate/molmut Community address: www.elsevier.com/locate/mutres Mutational and promoter hypermethylation status of FHIT gene in breast cancer patients of Kashmir 1 2 Nidda Syeed a,b,d , Syed Akhtar Husain b,d , A. Syed Sameer a,b , Q1 Nissar A. Chowdhri b,c , Mushtaq A. Siddiqi a,b,3 4 a Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, India 5 b Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, India 6 c Department of General Surgery, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, India 7 d Department of Biosciences, Jamia Millia Islamia, New Delhi, India 8 9 article info 10 11 Article history: 12 Received 3 September 2010 13 Received in revised form 2 November 2010 14 Accepted 12 November 2010 15 Available online xxx 16 Keywords: 17 FHIT 18 Breast cancer 19 Kashmir 20 Mutation 21 Hypermethylation 22 abstract Objective: Fragile histidine triad (FHIT) gene located at chromosome 3p14.2 is a putative tumor suppressor gene involved in the pathogenesis of breast cancer. Both genetic and epigenetic alterations in FHIT have been implicated in breast carcinoma. In the present study, our main aim was to study the impact of these two kinds of alterations of FHIT gene in breast cancer patients of Kashmir. Methods: We screened a total of 130 breast cancer patients of Kashmir by PCR-SSCP followed by direct sequencing and methylation specific PCR. Results: Mutational screening of FHIT gene revealed significant amount of mutations [40.7% (53/130)] in five hot spot exons (exons 5–9), FHIT promoter was found to be hypermethylated in 59 of 130 [45.3%] breast cancer patients in our population. Conclusion: In the present study we have shown a significant association between the mutational and hypermethylation profile of FHIT gene. Hence, we provide the first evidence to our knowledge that the significant association of FHIT mutation and hypermethylation leads to the complete inactivation of FHIT gene in patients with breast cancer. Silencing of the FHIT gene by promoter hypermethylation occurs in breast carcinomas, especially those with the significant amount of mutations. © 2010 Published by Elsevier B.V. 1. Introduction 23 Breast cancer is the most frequent neoplasm affecting women all 24 over the world and is third most common malignancy in the world 25 [1] with more than 1 million women diagnosed with breast can- 26 cer each year [2]. Breast cancer has been associated with a variety 27 of risk factors [3] both genetic and epigenetic changes [4]. Envi- 28 ronmental carcinogens have been shown to damage DNA at active 29 fragile sites by disrupting surveillance, which has been shown to 30 be tumorigenic [5,6]. Development of human breast cancer arises 31 from genetic alterations that drive the transformation of normal 32 mammary epithelial cells into highly malignant derivatives [7]. 33 The FHIT gene has been identified in the fragile locus, FRA3B, at 34 3p14.2 of human chromosome 3 [8], and was recently identified as 35 a candidate tumor suppressor gene [9]. FHIT gene is a member of the 36 histidine triad gene family, encoding a protein similar to the yeast 37 Corresponding author at: Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, India. Tel.: +91 194 2401013x2262; mobile: +91 9419767768; fax: +91 194 2403470. E-mail address: [email protected] (M.A. Siddiqi). diadenosine oligophosphates (ApnA) hydrolase, which are intra- 38 cellular and extracellular signaling molecules involved in cellular 39 differentiation and apoptosis. 40 DNA methylation has also been shown to contribute to car- 41 cinogenesis; hypermethylation of regulatory regions of many 42 tumor suppressor genes has been correlated with decreased gene 43 expression [10,11]. In previous studies, FHIT gene promoter hyper- 44 methylation has been correlated with loss of gene expression in 45 oesophageal, lung, breast, prostate, bladder, cervical, and oral can- 46 cers [12–17]. A recent study [18] has shown that methylation 47 profiles in familial breast cancer can be defined by the mutational 48 status that is different from the intrinsic subtypes. 49 FHIT, through binding or hydrolyzing Ap3A, influences the 50 Ap4A/Ap3A ratio, which is known to cause changes in cell prolif- 51 eration or to induce apoptosis [9,23]. However, controversy exists 52 with regard to a tumor suppressor role for FHIT [24,25]. Breast car- 53 cinoma emerges through a multistep process of transformation 54 of normal epithelium through hyperplasia, atypical hyperplasia, 55 and carcinoma in situ [26]. Loss of expression of a tumor sup- 56 pressor gene is also an important step in tumor progression from 57 premalignant, to in situ, to invasive carcinoma. Because a loss of 58 heterozygosity of 3p loci has been frequently observed in various 59 0027-5107/$ – see front matter © 2010 Published by Elsevier B.V. doi:10.1016/j.mrfmmm.2010.11.001

Transcript of Mutational and promoter hypermethylation status of FHIT gene in breast cancer patients of Kashmir

G

M

1

Mp

1

2

NQ1

N3

4

a5b6c7d8

9

a10

11

A12

R13

R14

A15

A

16

K17

F18

B19

K20

M21

H22

123

24

o25

[26

c27

o28

r29

f30

b31

f32

m33

34

335

a36

h37

SI

0d

ARTICLE IN PRESSModel

UT 10993 1–8

Mutation Research xxx (2010) xxx–xxx

Contents lists available at ScienceDirect

Mutation Research/Fundamental and MolecularMechanisms of Mutagenesis

journa l homepage: www.e lsev ier .com/ locate /molmutCommuni ty address : www.e lsev ier .com/ locate /mutres

utational and promoter hypermethylation status of FHIT gene in breast canceratients of Kashmir

idda Syeeda,b,d, Syed Akhtar Husainb,d, A. Syed Sameera,b,issar A. Chowdhrib,c, Mushtaq A. Siddiqia,b,∗

Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, IndiaDepartment of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, IndiaDepartment of General Surgery, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir, IndiaDepartment of Biosciences, Jamia Millia Islamia, New Delhi, India

r t i c l e i n f o

rticle history:eceived 3 September 2010eceived in revised form 2 November 2010ccepted 12 November 2010vailable online xxx

eywords:HIT

a b s t r a c t

Objective: Fragile histidine triad (FHIT) gene located at chromosome 3p14.2 is a putative tumor suppressorgene involved in the pathogenesis of breast cancer. Both genetic and epigenetic alterations in FHIT havebeen implicated in breast carcinoma. In the present study, our main aim was to study the impact of thesetwo kinds of alterations of FHIT gene in breast cancer patients of Kashmir.Methods: We screened a total of 130 breast cancer patients of Kashmir by PCR-SSCP followed by directsequencing and methylation specific PCR.Results: Mutational screening of FHIT gene revealed significant amount of mutations [40.7% (53/130)] in

reast cancerashmirutationypermethylation

five hot spot exons (exons 5–9), FHIT promoter was found to be hypermethylated in 59 of 130 [45.3%]breast cancer patients in our population.Conclusion: In the present study we have shown a significant association between the mutational andhypermethylation profile of FHIT gene. Hence, we provide the first evidence to our knowledge that thesignificant association of FHIT mutation and hypermethylation leads to the complete inactivation of FHITgene in patients with breast cancer. Silencing of the FHIT gene by promoter hypermethylation occurs in

ially t

38

39

40

41

42

43

44

45

46

47

48

49

breast carcinomas, espec

. Introduction

Breast cancer is the most frequent neoplasm affecting women allver the world and is third most common malignancy in the world1] with more than 1 million women diagnosed with breast can-er each year [2]. Breast cancer has been associated with a varietyf risk factors [3] both genetic and epigenetic changes [4]. Envi-onmental carcinogens have been shown to damage DNA at activeragile sites by disrupting surveillance, which has been shown toe tumorigenic [5,6]. Development of human breast cancer arisesrom genetic alterations that drive the transformation of normal

ammary epithelial cells into highly malignant derivatives [7].

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

The FHIT gene has been identified in the fragile locus, FRA3B, atp14.2 of human chromosome 3 [8], and was recently identified ascandidate tumor suppressor gene [9]. FHIT gene is a member of theistidine triad gene family, encoding a protein similar to the yeast

∗ Corresponding author at: Department of Immunology and Molecular Medicine,her-I-Kashmir Institute of Medical Sciences, Soura, Srinagar 190011, Kashmir,ndia. Tel.: +91 194 2401013x2262; mobile: +91 9419767768; fax: +91 194 2403470.

E-mail address: [email protected] (M.A. Siddiqi).

50

51

52

53

54

027-5107/$ – see front matter © 2010 Published by Elsevier B.V.oi:10.1016/j.mrfmmm.2010.11.001

hose with the significant amount of mutations.© 2010 Published by Elsevier B.V.

diadenosine oligophosphates (ApnA) hydrolase, which are intra-cellular and extracellular signaling molecules involved in cellulardifferentiation and apoptosis.

DNA methylation has also been shown to contribute to car-cinogenesis; hypermethylation of regulatory regions of manytumor suppressor genes has been correlated with decreased geneexpression [10,11]. In previous studies, FHIT gene promoter hyper-methylation has been correlated with loss of gene expression inoesophageal, lung, breast, prostate, bladder, cervical, and oral can-cers [12–17]. A recent study [18] has shown that methylationprofiles in familial breast cancer can be defined by the mutationalstatus that is different from the intrinsic subtypes.

FHIT, through binding or hydrolyzing Ap3A, influences theAp4A/Ap3A ratio, which is known to cause changes in cell prolif-eration or to induce apoptosis [9,23]. However, controversy existswith regard to a tumor suppressor role for FHIT [24,25]. Breast car-cinoma emerges through a multistep process of transformation

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

of normal epithelium through hyperplasia, atypical hyperplasia, 55

and carcinoma in situ [26]. Loss of expression of a tumor sup- 56

pressor gene is also an important step in tumor progression from 57

premalignant, to in situ, to invasive carcinoma. Because a loss of 58

heterozygosity of 3p loci has been frequently observed in various 59

ARTICLE IN PRESSG Model

MUT 10993 1–8

2 N. Syeed et al. / Mutation Research xxx (2010) xxx–xxx

5–9 sh

c60

s61

c62

p63

[64

65

q66

c67

p68

i69

70

a71

K72

v73

274

275

76

I77

i78

n79

80

f81

c82

w83

T84

C85

286

87

u88

w89

U90

291

92

F93

f94

r95

fi96

o97

I98

g99

r100

a101

U102

103

a104

w105

a106

i107

g108

b109

g110

v111

t112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

3. Results 139

Our study comprised of 130 breast cancer patients, revealed that 140

out of 130 cases 53 [40.7%] were mutated in one or more hot spot 141

Fig. 1. SSCP analysis of FHIT gene exons

ancers including breast cancer [27–32] recently, abnormal tran-cription of the FHIT gene has been reported in human breast cancerell lines and in primary breast cancers, showing that about 30% ofrimary breast cancers exhibited abnormalities in FHIT transcripts33].

It further seems to be necessary to examine not only the fre-uency of abnormalities of FHIT transcription in primary breastancers but also the association of the abnormalities with clinico-athological characteristics of cancer, which will be of great help

n characterizing the role of the FHIT gene in breast carcinogenesis.Therefore we devised this study to analyze the frequency of FHIT

berrations – both mutations and hypermethylation in our ethnicashmir Population and also to correlate the FHIT gene status witharious clinicopathological characteristics.

. Materials

.1. Patients and tumor tissue procurement

A cohort of 130 randomly selected breast cancer patients admitted to the Sher--Kashmir Institute of Medical Sciences was included in the study. All patientsncluded in the study were both male and female, with the histopathological diag-osis of the breast done under Institutional histopathology department.

Tissue samples consisting of tumor and adjacent normal were collected directlyrom Operation Theater of Department of General Surgery. Only histopathologi-al confirmed tumor tissues were included in the study. Peripheral blood samplesere also collected from the patients before the surgical resection of the tumor.

he study was approved by the Sher-I-Kashmir Institute of Medical Sciences Ethicalommittee.

.2. DNA isolation

Genomic DNA was extracted from tissue samples and peripheral blood samplessing DNA Extraction Kit (Qiagen, USA). The quality of the resulting genomic DNAas stringently assessed by low percentage agarose gel electrophoresis followed byV spectrophotometer.

.3. Conventional PCR-SSCP

In our study we screened the mutations in the open reading frame of theHIT gene, i.e., exons 5–9. Primer designs of these exons (5th to 9th) were takenrom earlier studied reports [35,36]. The DNA samples were amplified for targetegions/exons by conventional PCR. Each PCR reaction was performed in a 50 �lnal volume containing 20–100 ng of genomic DNA, 10 mmol/l Tris–HCl, 10 pmol/lf each primer, 200 �mol/l of each dNTP, and 1 U of Taq DNA polymerase (Genei,ndia). PCR was performed in a thermal cycler (Biorad icycler) using different pro-rams for five exons. Both positive and negative controls were used in each PCReaction. Negative controls were without DNA template to exclude non-specificmplification. The PCR products were run on 1.8% agarose gel and analyzed underV illuminator.

The high-quality amplified products were assessed with SSCP analysis. For thisssessment, 8 �l of each PCR product was added 2 �l of loading solution. The tubeas immediately heat-denatured at 95 ◦C for 5 min, then cooled on ice for 5 min, and

t 4 ◦C for 5 min, respectively, and kept there or on ice until a sample was loaded

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

nto the gel. 4 �l of each processed PCR product were electrophoresed on 1× MDETM

els. The products were run at 600 V for 10–12 h at room temperature in 1× TBEuffer solution (89 mM tris-base, 89 mM boric acid, 2 mM EDTA, and pH 8.0). Theels were stained using a non-radioactive silver staining method and results wereisualised and photographed with an image analyzer, as shown in Fig. 1. Sampleshat showed one or two band-shifts separated from the wild-type bands were iden-

owing mobility shifts in tumor sample.

tified as SSCP positive. All the samples that contained mutations were subjected tothe SSCP analysis procedure at least twice to rule out contamination.

2.4. Sequencing

The samples which showed variant band-shifts in SSCP were re-amplified forsequencing. The PCR products were gel-extracted using a Gel Extraction Kit (Qiagen,USA) and then sent for direct DNA sequencing. DNA sequencing was carried out atMacrogen Inc., Korea. To minimize the sequencing artifacts by PCR, products fromat least two different PCRs were sequenced using forward and reverse primers.

2.5. Methylation-specific polymerase chain reaction

Genomic DNA isolated from tumor and adjacent normal tissues by the proto-col described above was bisulphite modified by using commercial kit (MethylationDirect Kit, Zymo Research) according to manufacturer’s instructions. The modi-fied DNA were amplified by using methylation/unmethylation specific primers asreported earlier [37], 3 �l of bisulphite modified DNA was used in the PCR mixcontaining 1× PCR buffer, 200 �mol/l of each dNTP and 2 U Amp gold Taq DNApolymerase, 0.4 �M of primer and were amplified using the following reaction con-ditions: initial denaturation at 95 ◦C for 5 min followed by 38 cycles at 95 ◦C 50 s,62 ◦C 50 s, 72 ◦C 1 min, with the final incubation at 72 ◦C for 7 min. Ten-�l of the PCRproducts were loaded onto 6% denaturing polyacyrlamide gels and stained usingethidium bromide and visualised under UV illuminator, the PCR generated a 74 bpproduct both for methylated and unmethylated, as shown in Fig. 2.

2.6. Statistical analysis

Pearson’s two proportions test was used to compare the determined FHIT genemutational and methylational status with various clinical parameters. Differenceswith P > 0.05 were accepted as statistically not significant. Calculations were doneusing SPSS for Windows, version 11.5 (SPSS, Chicago, IL, USA).

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

Fig. 2. Methylation status of p16 and FHIT genes in normal and tumor tissues fromthe same patient (74 bp). Lane marked N: normal breast tissue, T: tumor tissue. Lanemarked M: methylated, lane marked U: unmethylated. Lane L: 100 bp molecularladder. Where T1 represents Bca3 and N1 is its adjacent normal tissue, T2 representsBca7 and N2 is its adjacent normal tissue. T3 represents Bca3.

ING

M

Resea

e142

o143

i144

3145

146

o147

(148

w149

1150

i151

a152

153

m154

o155

7156

i157

1158

e159

160

m161

c162

o163

(164

165

p166

6167

9168

p169

170

p171

a172

w173

174

o175

t176

p177

t178

w179

p180

3181

182

s183

s184

w185

s186

i187

t188

3189

190

s191

h192

n193

194

F195

m196

3197

198

t199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

ARTICLEModel

UT 10993 1–8

N. Syeed et al. / Mutation

xons of FHIT gene, and hypermethylation of the promoter regionf FHIT gene was found in 45.3% [59 of 130] of breast cancer casesn our population.

.1. Mutational analysis of FHIT in breast carcinomas

Mutational screening of the five hot spot exons (exons 5–9)f FHIT gene revealed significant amount of mutations [40.7%53/130)] in our population. In the module of direct sequencing, weere able to detect 53 mutations out of which 26 were missense,

3 were frameshifts, 7 were nonsense, and 7 were silent mutations,n addition to these we were also able to detect 10 novel mutations,s shown in Fig. 3.

Among the 26 missense mutations, there were six missenseutations present in exon-5 (three in codon 34, two in codon 24,

ne in codon 30), five in exon-6 (two in codon 82, two in codon7 and one in codon 74), four in exon-7 (two in codon 90 and two

n codon 89), eleven in exon-8 (two in codon 112, two in codon16 and one in codon 98, 103, 115, 102, 107, 110, 113) and none inxon-9.

Among the 13 frameshift mutations, there were six frameshiftutations in exon-5 (three in codon 20, two in codon 30 and one in

odon 31), four in exon-6 (two in codon 74 and two in codon 69),ne in exon-7 (one in codon 90), none in exon-8 and two in exon-9both in codon 146).

Among the 7 nonsense mutations, no nonsense mutation wereresent in exon-5, three were present in exon-6 (three in codon4), two were present in exon-7 (one in codon 93 and one in codon0), two were present in exon-8 (both in codon 103) and none wereresent in exon-9.

Among the 7 silent mutations, three silent mutations wereresent in exon-5 (three in codon 11), none were present in exon-6nd exon-7, three were present in exon-8 (three in codon 98), oneas present in exon-9 (one in codon 135).

The mutational analysis revealed that most of the mutationsccurred in exon 8 followed by exons 5–7, and very few muta-ions were found in exon-9. Most of the missense mutations wereresent in exon-8, a good number of frameshift and silent muta-ions were present in exon-5, large number of nonsense mutationsas present in exon-6, and most of the silent mutations wereresent in exon-8 as shown in Table 1.

.2. Frequency of FHIT hypermethylation in breast carcinomas

In the present study a total of 130 breast cancer patients weretudied for the methylational status of FHIT. Methylational analy-is of the breast tumors revealed that 45.3% (59/130) of FHIT geneere methylated. FHIT gene was also observed to be methylated in

ome adjacent normal tissues. Using statistical analysis, we exam-ned methylation with regard to clinicopathological parameters ofhese cancer patients.

.3. Clinicopathological association

In the present study, breast tumors carrying the mutations weretatistically significant with as menopausal status, nodal status,istopathological grade and breast involved. However there waso significant association with age, sex, and smoking status.

Breast tumors that were found to be hypermethylated forHIT gene were significantly associated with parameters like age,enopausal status, smoking status, nodal status and tumor stage.

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

.4. FHIT mutation and hypermethylation status: two hit

In 30 cancer samples there were both – inactivation of FHITumor suppressor gene by mutation and epigenetic silencing

PRESSrch xxx (2010) xxx–xxx 3

by promoter hypermethylation, leading to complete inacti-vation of FHIT, suggesting the involvement of both for theinactivation of FHIT that leads to tumor development and progres-sion.

4. Discussion

Breast cancer is one of the leading causes of death in femalesall over the world [1]. A number of alterations that occur at thegenetic level involving oncogenes, tumor suppressor genes as wellas DNA repair genes all of which are responsible for the devel-opment of breast carcinoma. But the complete inactivation of thetumor suppressor gene is highly affected by mutational and epige-netic silencing.

FHIT gene is a candidate tumor suppressor gene, since its dis-covery in 1996, alterations of the FHIT gene have been frequentlyobserved in a variety of tumors [8,38–41]. It has been postulatedthat in some cancers, including breast cancer, the alterations inthe FHIT gene represent an early event in carcinogenesis [42–45]whereas in other cancers aberrant FHIT transcripts occur at laterstages of cancer development [46].

4.1. Effect of the mutations of FHIT on breast cancer genetics

Present study evaluated the reported hot spot region of FHITgene (exon-5 to exon-9). A total of 40.7% tumors were found tobe mutated. The exon-8 which contains the histidine triad domainessential for anti cancer activity of FHIT protein housed most of themutations as has been reported earlier [47], followed by exon-7,exon-5 and exon-6. Exon-9 exhibited least number of mutations asis also reported in earlier studies [33]. The significant feature of thestudy is the seven nonsense mutations, that lead to the truncationof FHIT protein hence the reduced expression level of FHIT; when itsprotein is translated from transcripts lacking the non-coding exons3 or 4 [48].

Statistically significant association of FHIT gene mutations withcertain clinico-epidemiological characteristics, such as menopausalstatus, nodal status, histopathological grade and breast involvedwas observed as shown in Table 2. Significant association betweenFHIT mutation and pre-menopause stage suggests breast canceris more strongly associated with risk in premenopausal women,although the difference between pre- and post-menopausalwomen has not been reported to be statistically significant [49].Correlation of FHIT gene mutation with lymph node involve-ment has been found significant, suggesting its role in thebreast cancer metastases. To date axillary lymph node sta-tus is the best prognostic indicator in breast cancer patients[50,51]. Further, significant association of FHIT gene mutationwith histopathological grade has been found in the present study,suggesting that a distinctive mechanism is responsible to inac-tivate the tumor suppressor function of FHIT gene. Althoughwe found significant association of FHIT gene and the breastinvolved, but cannot assign any relevant reason about this asso-ciation.

4.2. Effect of FHIT hypermethylation related to breast cancerdevelopment

Aberrant promoter methylation changes that occur duringthe development of cancer are associated with transcriptionalrepression and loss of function of the gene by interrupting the

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

binding of proteins involved in transcription activator complex 255

[52]. 256

Hypermethylation of the FHIT promoter was detected in 59 of 257

the 130 (45.3%) cases examined. The presence of high percent- 258

age of hypermethylation in our case reveals that methylation is 259

ARTICLE IN PRESSG Model

MUT 10993 1–8

4 N. Syeed et al. / Mutation Research xxx (2010) xxx–xxx

F ve) alQ4a

o260

m261

c262

s263

ig. 3. Partial electropherograms representing the mutant (shown by arrows) (abond 8.

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

ne of the major determinants in tumor progression because inajority of the cases, expression of the genes in these breast

ancer cases correlates with hypermethylation of the promoterequences. The rate of hypermethylation is higher than the

ong with its normal sequence (below) of FHIT gene: (a) exons 5 and 6; (b) exons 7

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

percentage of mutation which suggests that FHIT hypermethy- 264

lation is a more common event in breast carcinoma. Promoter 265

aberrant methylation of FHIT is an important mechanism for 266

inactivation of this tumor suppressor gene in mammary tumori- 267

ARTICLE IN PRESSG Model

MUT 10993 1–8

N. Syeed et al. / Mutation Research xxx (2010) xxx–xxx 5

Table 1Details of FHIT mutations in exons 5–9 of breast cancer patients from Kashmir valley.Q5

Exon involved Codon number Base changeg Amino acid change Effecth

Exon 5 34 GGA > AGA Gly > Arg Missense11 AAG > AAA Lys > Lys Silent20 GAA > GAGC INS G Frameshift24 GCT > CCT Ala > Pro Missense11 AAG > AAA Lys > Lys Silent30 CCT > CAT Pro > His Missense30 CCT > CCTA INS A Frameshift34 GGA > AGA Gly > Arg Missense31 GTG > GTAG INS A Frameshift34 GGA > AGA Gly > Arg Missense20 GAA > GAGC INS G Frameshift24 GCT > CCT Ala > Pro Missense20 GAA > GAGC INS G Frameshift11 AAG > AAA Lys > Lys Silent30 CCT > CCTA INS A Frameshift

Exon 6 64 AGA > TGA Arg > Stop Nonsense82 ATG > AAG Met > Lys Missense74 CAT > CATA INS A Frameshift77 TCT > CCT Ser > Pro Missense69 GTG > GTAG INS A Frameshift64 AGA > TGA Arg > Stop Nonsense74 CAT > CAG His > Gln Missense69 GTG > GTAG INS A Frameshift77 TCT > CCT Ser > Pro Missense64 AGA > TGA Arg > Stop Nonsense82 ATG > AAG Met > Lys Missense74 CAT > CATA INS A Frameshift

Exon 7 90 CAG > CAT Gln > His Missense93 AAG > TAG Lys > Stop Nonsense89 GGA > AGA Gly > Arg Missense90 CAG > TAG Gln > Stop Nonsense90 CAG > CAT Gln > His Missense89 GGA > AGA Gly > Arg Missense90 CAG > CAAG INS A Frameshift

Exon 8 116 GAG > GGG Glu > Gly Missense98 CAT > CAC His > Lys Missense

103 AAG > AAA Arg > Lys Missense98 CAT > CAC His > His Silent

115 GAG > GTG Glu > Val Missense102 AGG > AAG Arg > Lys Missense103 AAG > TAG Lys > Stop Nonsense107 TTT > TCT Phe > Ser Missense

98 CAT > CAC His > His Silent110 AAT > AAG Asn > Lys Missense116 GAG > GTG Glu > Val Missense103 AAG > TAG Lys > Stop Nonsense112 AGC > ACC Ser > Thr Missense

98 CAT > CAC His > His Silent113 ATC > ACC Ile > Thr Missense112 AGC > ACC Ser > Thr Missense

> TT> GCC

> TT

g268

[269

t270

l271

m272

b273

s274

275

c276

[277

c278

m279

W280

t281

s282

283

284

285

286

287

288

289

290

291

292

293

Exon 9 146 TTT135 GCA146 TTT

enesis. Our results were similar to previously reported studies53,54]. We also found hypermethylation in some non-cancerousissues that might represent the appearance of premalignantesions. Thus, detection of FHIT hypermethylation using MSP

ight provide potential new molecular diagnostic markers ofreast carcinomas at an early stage during multistep carcinogene-is.

Clinicopathologically, aberrant FHIT expression has been asso-iated with pathogenesis and prognosis of various tumors8,34,55–61]. FHIT gene hypermethylation was found signifi-ant with some of the clinico-pathological parameters like age,

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

enopausal status, smoking status, nodal status and tumor stage.e found a significant association between FHIT gene hyperme-

hylation and age (>50 years) supported by enormous evidenceuggesting that some of the methylation changes may initi-

Del T FrameshiftAla > Ala SilentDel T Frameshift

ate in subpopulations of normal cells as a function of age andprogressively increase during carcinogenesis [62]. FHIT gene hyper-methylation was also found significant in pre-menopausal womensuggesting that the premenopausal women are at approximatelytwice risk, than the postmenopausal women, which has beenreported earlier that proliferation rate is somewhat lower andrelatively stable in post menopausal women [63–65]. Hyperme-thylation of the FHIT gene was associated with smoking variable;FHIT gene hypermethylation was significantly associated with eversmokers. As most of the female patients in our study originatedfrom far flung rural regions, where they have a tradition of Hukka

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

smoking (water pipe smoke) [66], which could be one of the possi- 294

ble reasons for hypermethylation. FHIT hypermethylation was also 295

found significantly associated with the lymph node involvement 296

and tumor stage (I and II). 297

ARTICLE IN PRESSG Model

MUT 10993 1–8

6 N. Syeed et al. / Mutation Research xxx (2010) xxx–xxx

Table 2Effect of FHIT mutation and hypermethylation pattern in the breast cancer patients from Kashmir valley.

Variable TotalN = 130 (%)

MutantsM = 53 (%)

OR; P-value; CI(95%)

MethylatedN = 59 (%)

OR; P-value; CI(95%)

SexMales 7 (53.8%) 4 (57.1%) 2.01; 0.44; 0.43–9.39 2 0.46; 0.45; 0.08–2.48Females 123 (94.6%) 49 (39.8%) 57Age>50 13 (10.0%) 8 2.56; 0.13; 0.78–8.31 11 7.90; 0.003; 1.68–37.28≤50 117 (90.0%) 45 48DwellingRural 45 (34.6%) 38 21.7; 0.000; 8.10–58.15 37 13.24; 5.47; 5.36–32.75Urban 85 (65.4%) 15 22Smoking statusNever 93 (71.5%) 38 0.89; 0.83; 0.39–2.03 35 0.33; 0.006; 0.15–0.72Ever 37 (28.5%) 15 24Menopausal statusPre 94 (72.3%) 21 2.71; 0.01; 1.23–5.96 21 2.25; 0.04;1.02–4.96Post 94 (72.3%) 32 38Nodal statusInvolved 34 (26.2%) 25 6.74; 0.000; 2.79–16.26 23 3.48; 0.002; 1.52–7.98Not Involved 96 (73.8%) 28 36Breast involvedR 35 (26.9%) 33 61.87; 0.000;

13.66–280.1230 13.65; 1.38; 4.81–38.73

L 95 (73.1%) 20 29Tumor stageII (a + b) 72 (55.4%) 35 2.10; 0.04; 1.01–4.33 26 0.43; 0.02; 0.21–0.86III (a + b) + IV 58 (44.6%) 18 30Histopathological tumor gradePD 25 (96.0%) 18 5.14; 0.0005;

1.96–13.4713 1.38; 0.50; 0.57–3.33

MD + WD 58 + 47 (24.1%) 18 + 17 27 + 19

Table 3Statistical relation between promoter hypermethylation and mutation of FHIT gene.

FHIT promoter hypermethylation versus FHIT mutation

FHIT methylation OR; P-value; CI(95%)

UnmethylationN = 71

MethylationN = 59

FHIT mutation2

3

4298

299

t300

i301

a302

h303

p304

i305

o306

n307

b308

i309

s310

m311

312

313

314

315

316

317

318

319

320

321

322

TC

Wild: N = 77 48

Mutant: N = 53 23

.3. Two hits inactivate the tumor suppressor role of FHIT

The important feature of our study was the significant associa-ion of FHIT mutation with promoter hypermethylation, as shownn Table 3. In 30 samples, we found both FHIT mutation as wells FHIT promoter hypermethylation. It has been reported that oneit cannot result in the complete loss of function of the tumor sup-ressor gene FHIT, and therefore two hits are required for complete

nactivation [67], where hypermethylation has been considered asne of the hits [21,68]. It is also known that methylation can occurot as a primary event, but secondary to mutation [20,68] as has

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

een reported [69] that methylation of tumor suppressors cannotnitiate tumorigenesis, because in order to control the gene expres-ion and/or methylation the methylator phenotype itself requiresutations in genes that control them.

able 4orrelation of tumor grade versus mutation versus hypermethylation status.

FHIT gene status Tumor grade

Low grade II

Double hit (N = 30) 7Single hit – mutation only (N = 23) 12Single hit – methylation only (N = 29) 15

9 0.46; 0.04; 0.22–0.94

0

Among these 30 samples most of the mutations were truncat-ing mutations, and when these truncating mutations were coupledwith methylation, there are many possibilities that the methy-lated allele has actually harboured a somatic or germ line mutationwhich has predisposed the gene to methylation. We found tumorgrade statistically significant with mutational and hypermethyla-tion status both (Table 4), suggesting that both of these aberrations(mutation + hypermethylation) together are responsible for theprogression of breast cancer to higher grade.

According to Tomlinson et al. [70] “two hits” hypothesis, oneallele of a tumor suppressor can be inactivated by methylation

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

when the other harbors a mutation or allelic loss. Our results also fit 323

this model, revealing a strong association between FHIT promoter 324

hypermethylation and the mutation, suggesting that in breast car- 325

cinomas, one allele is lost by mutation, and the other is inactivated 326

P-value; �2

High grade III + IV

2311 0.04; 6.4114

ING

M

Resea

b327

t328

F329

o330

i331

[332

5333

334

g335

m336

t337

s338

o339

m340

b341

e342

s343

C344

345

c346

A347

348

f349

S350

p351

c352

d353

fi354

UQ2355

356

A357

358

t359

r360

i361

s362

i363

364

w365

a366

c367

R368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

[ 393

394

[ 395

396

397

[ 398

399

400

[ 401

402

403

[ 404

405

406

407

408

[ 409

410

411

412

[ 413

414

415

[ 416

417

418

[ 419

420

421

[ 422

423

[ 424

425

[ 426

427

[ 428

429

430

[ 431

432

433

[ 434

435

436

[ 437

438

439

[ 440

441

442

[ 443

444

445

446

[ 447

448

449

[ 450

451

452

[ 453

454

455

456

[ 457

458

459

[ 460

461

462

ARTICLEModel

UT 10993 1–8

N. Syeed et al. / Mutation

y aberrant methylation, with both events leading simultaneouslyo the biallelic inactivation and complete lack of function of theHIT gene. In addition, it is presumed that methylation usuallyccurs monoallelic, and complete loss of expression of FHIT genes a consequence of a combination of methylation and mutation13].

. Conclusion

FHIT gene, tumor suppressor gene is involved in breast carcino-enesis and in various other malignancies. We have studied theutational and hypermethylation profile of FHIT, which reveals

hat it is associated with breast cancer development and progres-ion. Our study reveals that the genetic and epigenetic aberrationsf FHIT gene lead to its complete inactivation, which leads to mam-ary carcinogenesis. The clinical significance of our findings should

e further evaluated in larger cohorts with longer follow-up tovaluate the effect of these findings on disease-free and overallurvival.

onflict of interest

The contributing authors have no financial or any non-financialompeting interests.

uthors’ contributions

Nidda Syeed formulated, designed and performed the lab workor the study. Akhtar Hussain supervised the analysis, A. Syedameer helped in the lab work. Nissar A. Chowdhri procured androvided the tumor samples for the study and Mushtaq A. Siddiqioordinated the study, revised the manuscript and entire work wasone under his supervision. All authors have read and approved thenal manuscript.

ncited references

[19,22].

cknowledgements

The authors gratefully acknowledge the Sher-I-Kashmir Insti-ute of Medical Sciences, Kashmir for providing funds for thisesearch work. The present research was not funded by any fund-ng agency. The collection of cancer samples used in this study wasupported by the Department of General Surgery, Sher-I-Kashmirnstitute of Medical Sciences.

The authors would like to thank all the breast cancer patientsho participated in the study who are responsible for the creation

nd maintenance of the entire group within which this study wasonducted but were not involved in the current paper.

eferences

[1] S. Sipetic, V. Petrovic, Z. Milic, H. Vlajinac, Breast cancer incidence amongwomen of Branicevo region in the period 1991–2000, Med. Pregl. 57 (2004)467–472.

[2] D.M. Parkin, F. Bray, J. Ferlay, P. Pisani, Global cancer statistics, 2002, CA CancerJ. Clin. 55 (2005) 74–108.

[3] R. Kaaks, S. Rinaldi, T.J. Key, F. Berrino, P.H. Peeters, C. Biessy, et al., Post-menopausal serum androgens, oestrogens and breast cancer risk: the Europeanprospective investigation into cancer and nutrition, Endocr. Relat. Cancer 12

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

(2005) 1071–1082.[4] G.S. Xie, A.R. Hou, L.Y. Li, Y.N. Gao, S.J. Cheng, Aberrant p16 promoter hyperme-

thylation in bronchial mucosae as a biomarker for the early detection of lungcancer, Chin. Med. J. 119 (2006) 1469–1472.

[5] S. Gasser, D. Raulet, The DNA damage response, immunity and cancer, Semin.Cancer Biol. 16 (2006) 344–347.

[

[

PRESSrch xxx (2010) xxx–xxx 7

[6] M.F. Arlt, A.M. Casper, T.W. Glover, Common fragile sites, Cytogenet. GenomeRes. 100 (2003) 92–100.

[7] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70.[8] M. Ohta, H. Inoue, M.G. Cotticelli, K. Kastury, R. Baffa, J. Palazzo, et al., The

FHIT gene, spanning the chromosome 3p14.2 fragile site and renal carcinoma-associated t(3;8) breakpoint, is abnormal in digestive tract cancers, Cell 84(1996) 587–597.

[9] L.L. Kisselev, J. Justensen, A.D. Wolfson, L.Y. Frolova, Diadenosine oligophos-phates (ApnA), a novel class of signaling molecules? FEBS Lett. 427 (1998)157–163.

10] M. Esteller, P.G. Corn, S.B. Baylin, J.G. Herman, A gene hypermethylation profileof human cancer, Cancer Res. 61 (2001) 3225–3229.

11] M.L. Gonzalgo, T. Hayashida, C.M. Bender, et al., The role of DNA methylationin expression of the p19/p16 locus in human bladder cancer cell lines, CancerRes. 58 (1998) 1245–1252.

12] H. Tanaka, Y. Shimada, H. Harada, et al., Methylation of the 50 CpG Island of theFHIT gene is closely associated with transcriptional inactivation in esophagealsquamous cell carcinomas, Cancer Res. 58 (1998) 3429–3434.

13] R.A. Naqvi, A. Hussain, M. Raish, et al., Specific 50CpG island methylation signa-tures of FHIT and p16 genes and their potential diagnostic relevance in Indianbreast cancer patients, DNA Cell Biol. 27 (2008) 517–525.

14] R. Maruyama, S. Toyooka, K.O. Toyooka, K. Harada, A.K. Virmani, S. Zochbauer-Muller, A.J. Farinas, F. Vakar-Lopez, J.D. Minna, A. Sagalowsky, B. Czerniak,Gazdar AF, Aberrant promoter methylation profile of bladder cancer andits relationship to clinicopathological features, Cancer Res. 61 (2001)8659–8663.

15] R. Maruyama, S. Toyooka, K.O. Toyooka, A.K. Virmani, S. Zochbauer-Muller, A.J.Farinas, J.D. Minna, J. McConnell, E.P. Frenkel, AF. Gazdar, Aberrant promotermethylation profile of prostate cancers and its relationship to clinicopatholog-ical features, Clin. Cancer Res. 8 (2002) 514–519.

16] A.K. Virmani, C. Muller, A. Rathi, S. Zoechbauer-Mueller, M. Mathis, A.F. Gazdar,Aberrant methylation during cervical carcinogenesis, Clin. Cancer Res. 7 (2001)584–589.

17] K.W. Chang, S.Y. Kao, R.J. Tzeng, C.J. Liu, A.J. Cheng, S.C. Yang, Y.K. Wong, SC.Lin, Multiple molecular alterations of FHIT in betel-associated oral carcinoma,J. Pathol. 196 (2002) 300–306.

18] J.M. Flanagan, S. Cocciardi, N. Waddell, DNA methylome of familial breast canceridentifies distinct profiles defined by mutation status, Am. J. Hum. Genet. 86(2010) 420–433.

19] K.D. Robertson, P.A. Jones, DNA methylation: past, present and future direc-tions, Carcinogenesis 21 (2000) 461–467.

20] R.E. Watson, J.I. Goodman, Epigenetics, DNA methylation come of age in toxi-cology, Toxicol. Sci. 67 (2002) 11–16.

21] S.B. Baylin, J.G. Herman, J.R. Graff, et al., Alterations in DNA methylation: afundamental aspect of neoplasia, Adv. Cancer Res. 72 (1998) 141–196.

22] J.F. Costello, M.C. Fruhwald, D.J. Smiraglia, et al., Aberrant CpG-island methyla-tion has non-random and tumour-type specific patterns, Nat. Genet. 24 (2000)132–138.

23] A. Vartanian, I. Prudovsky, H. Suzuki, I. Dal Pra, L. Kisselev, Opposite effects ofcell differentiation and apoptosis on Ap3A/Ap4A ratio in human cell cultures,FEBS Lett. 415 (1997) 160–162.

24] J.M. Ordway, M.A. Budiman, Y. Korshunova, Identification of novel high-frequency DNA methylation changes in breast cancer, PLoS One 2 (12) (2007)e1314.

25] G.A. Otterson, G.H. Xiao, J. Geradts, F. Jin, W. Chen, W. Niklinska, et al., Proteinexpression and functional analysis of the FHIT gene in human tumor cells, J.Natl. Cancer Inst. 90 (1998) 426–432.

26] M.W. Beckmann, D. Niederacher, H.G. Schnurch, B.A. Gusterson, H.G. Bender,Multistep carcinogenesis of breast cancer and tumor heterogeneity, J. Mol. Med.75 (1997) 429–439.

27] E. Tsuchiya, Y. Nakamura, S.-Y. Weng, K. Nakagawa, S. Tsuchiya, H. Sugano,T. Kitagawa, Allelotype of non-small cell lung carcinoma: comparison betweenloss of heterozygosity in squamous cell carcinoma and adenocarcinoma, CancerRes. 52 (1992) 2478–2481.

28] S. Yokoyama, K. Yamakawa, E. Tsuchiya, M. Murata, S. Sakiyama, Y. Nakamura,Deletion mapping on the short arm of chromosome3 in squamous cell carci-noma and adenocarcinoma, Cancer Res. 52 (1992) 873–877.

29] R. Morita, J. Ishikawa, M. Tsutsumi, K. Hikiji, Y. Tsukada, S. Kamidono, S.Maeda, Y. Nakamura, Allelotype of renal cell carcinoma, Cancer Res. 51 (1991)820–823.

30] J. Yokota, Y. Tsukada, T. Nakajima, M. Gotoh, Y. Shimosato, N. Mon, Y.Tsunokawa, T. Sugimura, M. Terada, Loss of heterozygosity on the short armof chromosome 3 in carcinoma of the uterine cervix, Cancer Res. 49 (1989)3598–3601.

31] T. Sato, F. Akiyama, G. Sakamoto, F. Kasumi, Y. Nakamura, Accumulation ofgenetic alterations and progression of primary breast cancer, Cancer Res. 51(1991) 5794–5799.

32] L.-C. Chen, K. Matsumura, G. Deng, W. Kurisu, B.-M. Ljung, M.I. Lerman, F.M.Waldman, H.S. Smith, Deletion of two separate regions on chromosome3p inbreast cancers, Cancer Res. 54 (1994) 3021–3024.

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

33] M. Negrini, C. Monaco, I. Vorechovsky, M. Ohta, 1. Druck, R. Baffa, Huebner K., 463

M.C. Croce, The FHIT gene at 3pl4.2 is abnormal in breast carcinomas, Cancer 464

Res. 56 (1996) 3173–3179. 465

34] M.P. Madigan, R.G. Zieglar, J. Benichou, C. Byrne, R.N. Hoover, Proportion of 466

breast cancer cases in the United States explained by well-established risk 467

factors, J. Natl. Cancer Inst. 87 (1995) 1681–1685. 468

ING

M

8 Resea

[469

470

471

[472

473

474

[475

476

477

[478

479

480

481

[482

483

484

485

486

[487

488

489

[490

491

492

[493

494

495

[496

497

[498

499

500

[501

502

503

[504

505

506

[507

508

509

510

[511

512

513

514

[515

516

517

[518

519

[520

521

522

[523

[ 524

525

526

527

528

[ 529

530

531

[ 532

533

534

[ 535

536

537

[ 538

539

540

541

[ 542

543

544

545

[ 546

547

548

549

[ 550

551

552

553

[ 554

555

556

[ 557

558

[ 559

560

561

[ 562

563

564

[ 565

566

567

568

[ 569

570

571

[ 572

573

ARTICLEModel

UT 10993 1–8

N. Syeed et al. / Mutation

35] A. Gemma, K. Hagiwarn, Y. Ke, L.M. Burke, M.A. Khan, M. Nagashima, W.P. Ben-nett, C.C. Harris, FHIT mutations in human primary gastric cancer, Cancer Res.57 (1997) 1435–1437.

36] G. Cecener, Ü. Egeli, B. Tunca, I. Tasdelen, S. Tolunay, N. Bilgel, Importance ofnovel sequence alterations in the FHIT gene on formation of breast cancer,Tumori 93 (2007) 597–603.

37] A.O. Nygren, N. Ameziane, H.M. Duarte, et al., Methylation-specific MLPA (MS-MLPA): simultaneous detection of CpG methylation and copy number changesof up to 40 sequences, Nucleic Acids Res. 33 (14) (2005) e128.

38] J.D. Potter, J.R. Cerhan, T.A. Sellers, P.G. McGovem, C. Drinkard, L.R. Kushi, A.R.Folsom, Progesterone and estrogen receptors and mammary neoplasia in theIowa women’s health study: how many kinds of breast cancers are there?Cancer Epidemiol. Biomar. Prev. 4 (1995) 319–326.

39] K.M. Fong, E.J. Biesterveld, A. Virmani, I. Wistuba, Y. Sekido, S.A. Bader, M.Ahmadian, S.T. Ong, F.V. Rassool, P.V. Zimmerman, G. Giaccone, A.F. Gazdar,J.D. Minna, FHIT and FRA3B 3p14.2 allele loss are common in lung cancer andpreneoplastic bronchial lesions and are associated with cancer-related FHITcDNA splicing aberrations, Cancer Res. 57 (1997) 2256–2267.

40] S. Ingvarson, B.I. Sigbjornsdottir, C. Huiping, J.G. Jonasson, B.A. Agnarsson, Alter-ations of the FHIT gene in breast cancer: association with tumor progressionand patient survival, Cancer Detect. Prev. 25 (2001) 292–298.

41] B.Z. Yuan, C. Keck-Waggoner, D.B. Zimonjic, S.S. Thorgeirsson, N.C. Popescu,Alterations of the FHIT gene in human hepatocellular carcinoma, Cancer Res.60 (2000) 1049–1053.

42] R. Baffia, M.L. Veronese, B. Santoro, B. Mandes, J.P. Palazzo, M. Rugge, E. Santoro,C.M. Croce, K. Huebner, Loss of FHIT expression in gastric carcinoma, Cancer Res.58 (1998) 4708–4714.

43] M. Zou, Y. Shi, N.R. Farid, S.T. al-Sedairy, M.C. Paterson, FHIT gene abnormalitiesin both benign and malignant thyroid tumors, Eur. J. Cancer 35 (1999) 467–472.

44] Z. Guo, S.L. Johansson, J.S. Rhim, J.K. Vishwanatha, Fragile histidine triad geneexpression in primary prostate cancer and in an in vitro model, Prostate 43(2000) 101–110.

45] C. Huiping, J.G. Jonasson, B.A. Agnarsson, B.I. Sigbjornsdottir, K. Huebner, S.Ingvarsson, Analysis of the fragile histidine triad (FHIT) gene in lobular breastcancer, Eur. J. Cancer 36 (2000) 1552–1557.

46] K. Yoshino, T. Enomoto, T. Nakamura, H. Sun, K. Ozaki, R. Nakashima, H. Wanada,J. Saitoh, Y. Watanabe, K. Noda, Y. Murata, FHIT alterations in cancerous andnon-cancerous cervical epithelium, Int. J. Cancer 85 (2000) 6–13.

47] Z. Siprashvili, G. Sozzi, L.D. Barnes, P. McCue, A.K. Robinson, V. Eryomin, L. Sard,E. Tagliabue, A. Greco, L. Fusetti, G. Schwartz, M.A. Pierotti, C.M. Croce, K. Hueb-ner, Replacement of FHIT in cancer cells suppresses tumorigenicity, Proc. Natl.Acad. Sci. U.S.A. 94 (1997) 13771–13776.

48] G. Sozzi, S. Tornielli, E. Tagliabue, L. Sard, F. Pezzella, U. Pastorino, F. Minolitti,S. Pilotti, C. Ratcliffe, M.L. Veronese, P. Goldstraw, K. Huebner, C.M. Croce, MA.Pierotti, Absence of FHIT protein in primary lung tumors and cells lines withFHIT gene abnormalities, Cancer Res. 57 (1997) 5207–5212.

49] C. Nagata, N. Takatsuka, S. Inaba, N. Kawakami, H. Shimizu, Effect of soymilkconsumption on serum estrogen concentrations in premenopausal Japanesewomen, J. Natl. Cancer Inst. 90 (1998) 1830–1835.

50] C.L. Carter, C. Allen, D.E. Henson, Relation of tumor size, lymph node status, and

Please cite this article in press as: N. Syeed, et al., Mutational and promotKashmir, Mutat. Res.: Fundam. Mol. Mech. Mutagen. (2010), doi:10.1016/j

survival in 24,740 breast cancer cases, Cancer 63 (1989) 181–719.51] J. Russo, J. Frederick, H.E. Ownby, G. Fine, M. Hussain, H.I. Kirckstein, et al.,

Predictors of recurrence and survival of patients with breast cancer, Am. J. Clin.Pathol. 88 (1987) 123–131.

52] P.A. Jones, S.B. Baylin, The fundamental role of epigenetic events in cancer, Nat.Rev. Genet. 3 (2002) 415–428.

[

[

[

PRESSrch xxx (2010) xxx–xxx

53] M. Raish, V.S. Dhillon, A. Ahmad, M.A. Ansari, S. Mudassar, M. Shahid, V. Batra,P. Gupta, B.C. Das, N. Shukla, S.A. Husain, Promoter hypermethylation in tumorsuppressing genes p16 and FHIT and their relationship with estrogen receptorand progesterone receptor status in breast cancer patients from Northern India,Transl. Oncol. 4 (2009) 264–270.

54] J.C. Roa, L. Anabalón, O. Tapia, J. Martínez, J.C. Araya, M. Villaseca, P. Guzmán, I.E.Roa, Promoter methylation profile in breast cancer, Rev. Med. Chile 132 (2004)1069–1077.

55] Z. Gatalica, S.M. Lele, B.A. Rampy, B.A. Norris, The expression of FHIT proteinis related inversely to disease progression in patients with breast carcinoma,Cancer (Phila.) 88 (2000) 1378–1383.

56] M. Campiglio, Y. Pekarsky, S. Menard, E. Tagliabue, S. Pilotti, C.M. Croce, FHITloss of function in human primary breast cancer correlates with advanced stageof the disease, Cancer Res. 59 (1999) 3866–3869.

57] Q. Yang, G. Yoshimura, T. Suzuma, T. Tamaki, T. Umemura, M. Nakamura, Y.Nakamura, X. Wang, I. Mori, T. Sakurai, K. Kakudo, Clinicopathological sig-nificance of fragile histidine triad transcription protein expression in breastcarcinoma, Clin. Cancer Res. 7 (2001) 3869–3873.

58] D.L. Greenspan, D.C. Connolly, R. Wu, R.Y. Lei, J.T. Vogelstein, Y.T. Kim, J.E.Mok, N. Munoz, F.X. Bosch, K. Shah, K.R. Cho, Loss of FHIT expression in cer-vical carcinoma cell lines and primary tumors, Cancer Res. 57 (1997) 4697–4698.

59] S.I. Hayashi, K. Tanimoto, K. Hajiro-Nakanishi, E. Tsuchiya, M. Kurosumi, Y.Higashi, K. Imai, K. Suga, K. Nakachi, Abnormal FHIT transcripts in human breastcarcinomas: a clinicopathological and epidemiological analysis of 61 Japanesecases, Cancer Res. 57 (1997) 1981–1985.

60] G. Sozzi, M.L. Veronese, M. Negrini, R. Baffa, M.G. Cotticelli, H. Inoue, S. Tomielli,S. Pilotti, L. De Gregorio, U. Pastorino, M.A. Pierotti, M. Ohta, K. Huebner,C.M. Croce, The FHIT gene 3p14.2 is abnormal in lung cancer, Cell 85 (1996)17–26.

61] J.I. Lee, J.C. Soria, K. Hassan, D. Liu, X. Tang, A. El-Naggar, W.K. Hong, L. Mao, Lossof FHIT expression is a predictor of poor outcome in tongue cancer, Cancer Res.61 (2001) 837–841.

62] N. Ahuja, Q. Li, A.L. Mohan, S.B. Baylin, J.P. Issa, Aging and DNA methylation incolorectal mucosa and cancer, Cancer Res. 58 (1998) 5489–5494.

63] D.J. Ferguson, T.J. Anderson, Morphological evaluation of cell turnover in rela-tion to the menstrual cycle in the “resting” human breast, Br. J. Cancer 44 (1981)177–181.

64] T.A. Longacre, S.A. Bartow, A correlative morphologic study of human breastand endometrium in the menstrual cycle, Am. J. Surg. Pathol. 10 (6) (1986)382–393.

65] J.S. Meyer, Cell proliferation in normal human breast ducts, fibroadenomas, andother ductal hyperplasias measured by nuclear labeling with tritiated thymi-dine. Effects of menstrual phase, age, and oral contraceptive hormones, Hum.Pathol. 8 (1977) 67–81.

66] M.M. Mir, N.A. Dar, S. Gochhait, S.A. Zargar, A.G. Ahangar, R.N. Bamezai, p53Mutation profile of squamous cell carcinomas of the esophagus in Kashmir(India): a high-incidence area, Int. J. Cancer 116 (2005) 62–68.

67] A.G. Knudson Jr., H.W. Hethcote, B.W. Brown, Mutation and childhood cancer:a probabilistic model for the incidence of retinoblastoma, Proc. Natl. Acad. Sci.

er hypermethylation status of FHIT gene in breast cancer patients of.mrfmmm.2010.11.001

U.S.A. 72 (1975) 5116–5120. 574

68] P.A. Jones, P.W. Laird, Cancer epigenetics comes of age, Nat. Genet. 21 (1999) 575

163–167. 576

69] I. Tomlinson, M. Novelli, W. Bodmer, The mutation rate and cancer, Proc. Natl. 577

Acad. Sci. U.S.A. 93 (1996) 14800–14803. 578

70] I.P.M. Tomlinson, R. Roylance, R.S. Houlston. Q3 579