Oncotarget

Research Papers:

Genomic characterization of liver metastases from colorectal cancer patients

PDF |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:72908-72922. https://doi.org/10.18632/oncotarget.12140

Metrics: PDF 4390 views  |   HTML 4603 views  |   ?  

José María Sayagués, Luís Antonio Corchete, María Laura Gutiérrez, Maria Eugenia Sarasquete, María del Mar Abad, Oscar Bengoechea, Encarna Fermiñán, María Fernanda Anduaga, Sofia del Carmen, Manuel Iglesias, Carmen Esteban, María Angoso, Jose Antonio Alcazar, Jacinto García, Alberto Orfao _ and Luís Muñoz-Bellvis

Abstract

José María Sayagués1, Luís Antonio Corchete2, María Laura Gutiérrez1, Maria Eugenia Sarasquete2, María del Mar Abad3, Oscar Bengoechea3, Encarna Fermiñán4, María Fernanda Anduaga5, Sofia del Carmen3, Manuel Iglesias5, Carmen Esteban5, María Angoso5, Jose Antonio Alcazar5, Jacinto García5,*, Alberto Orfao1,*, Luís Muñoz-Bellvis5,*

1Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center, IBMCC-CSIC/USAL and IBSAL, University of Salamanca, Salamanca, Spain

2Cancer Research Center and Service of Hematology, University Hospital of Salamanca, Salamanca, Spain

3Department of Pathology, University Hospital of Salamanca, Salamanca, Spain

4Genomics Unit, Cancer Research Center, IBMCC-CSIC/USAL, Salamanca, Spain

5Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain

*These authors contributed equally to this work

Correspondence to:

Alberto Orfao, email: [email protected]

Keywords: GEP, colorectal cancer

Received: April 06, 2016     Accepted: September 13, 2016     Published: September 20, 2016

ABSTRACT

Metastatic dissemination is the most frequent cause of death of sporadic colorectal cancer (sCRC) patients. Genomic abnormalities which are potentially characteristic of such advanced stages of the disease are complex and so far, they have been poorly described and only partially understood. We evaluated the molecular heterogeneity of sCRC tumors based on simultaneous assessment of the overall GEP of both coding mRNA and non-coding RNA genes in primary sCRC tumor samples from 23 consecutive patients and their paired liver metastases. Liver metastases from the sCRC patients analyzed, systematically showed deregulated transcripts of those genes identified as also deregulated in their paired primary colorectal carcinomas. However, some transcripts were found to be specifically deregulated in liver metastases (vs. non-tumoral colorectal tissues) while expressed at normal levels in their primary tumors, reflecting either an increased genomic instability of metastatic cells or theiradaption to the liver microenvironment. Newly deregulated metastatic transcripts included overexpression of APOA1, HRG, UGT2B4, RBP4 and ADH4 mRNAS and the miR-3180-3p, miR-3197, miR-3178, miR-4793 and miR-4440 miRNAs, together with decreased expression of the IGKV1-39, IGKC, IGKV1-27, FABP4 and MYLK mRNAS and the miR-363, miR-1, miR-143, miR-27b and miR-28-5p miRNAs. Canonical pathways found to be specifically deregulated in liver metastatic samples included multiple genes related with intercellular adhesion and the metastatic processes (e.g., IGF1R, PIK3CA, PTEN and EGFR), endocytosis (e.g., the PDGFRA, SMAD2, ERBB3, PML and FGFR2), and the cell cycle (e.g., SMAD2, CCND2, E2F5 and MYC). Our results also highlighted the activation of genes associated with the TGFβ signaling pathway, -e.g. RHOA, SMAD2, SMAD4, SMAD5, SMAD6, BMPR1A, SMAD7 and MYC-, which thereby emerge as candidate genes to play an important role in CRC tumor metastasis.


INTRODUCTION

Occurrence of distant metastases is the main cause of sporadic colorectal cancer (sCRC) death, and the liver is the most common site for metastatic spread of the primary tumor [1, 2]. Metastasis is a complex multi-step process leading to the accumulation of genomic alterations in single tumor cells over the lifetime of a tumor from a benign lesion to invasive and metastatic spreading states leading to patient death [3, 4]. Genomic abnormalities which are potentially characteristic of such advanced stages of the disease are complex and so far, they have been poorly described and only partially understood. This relates to the fact that most genomic studies performed in colorectal cancer have focused on primary tumors, particularly in stage II disease, at diagnosis; in contrast, few studies have compared the deregulated transcripts of primary versus paired metastatic samples. Despite this, multiple mRNAs and miRNAS found to be expressed in primary tumors have been associated with metastatic colorectal carcinoma. Among others, these include mRNA of PTEN/PI3K [5], EGFR [6], TGFβ [7], and TP53 [8], as well as the metastatic CRC-associated miRNAs, miR-31 [9], miR-503 [10] and miR-133a [11].

Microarray analysis allows simultaneous investigation of several thousands of cancer-related and cancer-specific genes [12], such gene expression profiling (GEP) being highlighted as a potential tool to identify biomarkers for better prognostication and treatment of different types of cancer, including sCRC [13]. Thus, microarray data might provide significant insight into the biological differences between patients within a given cancer type with good- and poor-prognosis, at the same time it might be used as a screening tool for the identification of molecules to be targeted by existing or future (individualized) therapies. In recent years, several GEP have been identified as predictors for CRC stage II patient outcome. These include those being tested with the Oncotype DX® Colon Cancer test (Genomic Health, Inc., Redwood City, CA) [14] and Coloprint® (Agendia, Inc., Irvine, CA) gene chips, among other GEP platforms [13]. However, the molecular mechanisms underlying the association of such genomic profiles with metastatic colorectal carcinoma remain largely unknown.

Here we evaluate the molecular heterogeneity of sCRC tumors based on simultaneous assessment of the overall GEP for both coding mRNA and non-coding RNA genes -including miRNA, small nucleolar and large intergenic RNAs- in primary sCRC tumor samples from 23 consecutive patients and their paired liver metastases vs. non-tumoral tissues (n = 9). Overall, our results define a common GEP for all metastatic sCRC, which confirms and extends on previous observations [15] revealing new (specific) mRNA and miRNA signatures to be (potential) protective biomarker signatures for distant-disease.

RESULTS

Overall transcriptional profile of metastatic sCRC

Venn diagram analysis showed a total of 9,157 mRNA (3,744 down- and 5,413 up-regulated genes) and 118 miRNA genes (60 down- and 58 up-regulated) to be deregulated in primary tumors (vs. non-tumoral colorectal tissue) and 10,454 mRNA (4,777 down- and 5,677 up-regulated) and 254 miRNA genes (68 down- and 186 up-regulated) to be altered in liver metastases (vs. non-tumoral colorectal tissue). Overall, primary tumors and their paired liver metastases shared a total of 7,339 mRNA (3,297 down- and 4,042 up-regulated) and 109 miRNA (53 down- and 56 up-regulated) deregulated genes (Figure 1). Interestingly, one quarter of all mRNA (1,815/9,157; 20%) and miRNA transcripts (15/118; 13%) was exclusively found in primary tumors while one third of all differentially expressed mRNA transcripts (3,021/10,454; 29%) and the majority miRNA transcripts (145/254; 57%) were found to be deregulated only in liver metastases. Those genes most strongly overexpressed in primary tumors included the FOXQ1, MMP7, CLDN1, TACSTD2 and COL11A1 mRNAs and the miR-31, miR-4417, miR-503, miR-3647 and miR-592 miRNA transcripts; in turn, the CLCA4, CA1, SLC4A4, AQP8, and ZG16 mRNAs together with the miR-215, miR-139, miR-133a, miR-378c and miR-378d miRNAs were those genes showing the strongest down-regulated levels across all primary tumor samples analyzed (Table 1). Of note, those mRNAs and miRNAs transcripts differentially expressed in primary tumors vs. non-tumoral colorectal tissues, allowed for clear cut discrimination between both types of samples (Figure 2).

Gene expression profiles (GEP) of primary vs. metastatic colorectal cancer.

Figure 1: Gene expression profiles (GEP) of primary vs. metastatic colorectal cancer. (Panel A) Venn diagram representing mRNA and miRNA differentially expressed between the different types of samples analyzed: primary sporadic colorectal tumors vs. normal colorectal tissue, colorectal liver metastases vs. normal colorectal tissue and primary sporadic colorectal tumors vs. their paired liver metastases (q-values < 0.01). (Panel B) Most represented canonical pathways involved in metastatic colorectal tumors as defined by their GEP of both coding and non-coding RNAs (q < 0.01).

Table 1: Top mRNAs and miRNAs up- and down-regulated in primary sporadic colorectal tumors (n = 23) vs. non-tumoral colorectal tissues (n = 10)

Gene Name

Gene ID

Fold Change T vs. Non-T

Chr. band

Start (bp)

Stop (bp)

Strand

Transcript description

Up-regulated mRNA transcripts

FOXQ1

ENSG00000164379

44.1

6p25

1312440

1314758

+

protein-coding

MMP7

ENSG00000137673

31.1

11q21

102520508

102530753

protein-coding

CLDN1

ENSG00000163347

26.2

3q28

190305701

190322475

protein-coding

TACSTD2

ENSG00000184292

22.9

1p32

58575423

58577773

protein-coding

COL11A1

ENSG00000060718

21.4

1p21

102876467

103108496

protein-coding

KIAA1199

ENSG00000103888

17.1

15q24

80779343

80951776

+

protein-coding

CTHRC1

ENSG00000164932

14.7

8q22

103371515

103383005

+

protein-coding

DUSP27

ENSG00000198842

14.4

1q24

167094045

167129165

+

protein-coding

KRT23

ENSG00000108244

13.8

17q21

40922696

40937643

protein-coding

SRPX2

ENSG00000102359

12.3

Xq22

100644166

100671299

+

protein-coding

NFE2L3

ENSG00000050344

10.8

7p15

26152227

26187137

+

protein-coding

Up-regulated miRNA transcripts

hsa-miR-31

ENSG00000199177

10.3

9p21

21512114

21512184

hsa-miR

hsa-miR-4417

ENSG00000264341

8.2

1p36

5624131

5624203

+

hsa-miR

hsa-miR-503

ENSG00000208005

8.1

Xq26

133680358

133680428

hsa-miR

hsa-miR-1290

ENSG00000221662

5.9

1p34

18897071

18897148

hsa-miR

hsa-miR-3687

ENSG00000264063

5.5

21p11

9826203

9826263

+

hsa-miR

hsa-miR-592

ENSG00000207692

4.9

7q31

126698142

126698238

hsa-miR

hsa-miR-183

ENSG00000207691

4.6

7q32

129774905

129775014

hsa-miR

hsa-miR-224

ENSG00000207621

4.5

Xq28

151958578

151958658

hsa-miR

hsa-miR-1246

ENSG00000207584

4.2

2q31

176600980

176601052

hsa-miR

hsa-miR-21

ENSG00000199004

3.9

17q23

59841266

59841337

+

hsa-miR

hsa-miR-424

ENSG00000223749

3.8

Xq26

133677367

133680741

hsa-miR

Down-regulated mRNA transcripts

CLCA4

ENSG00000016602

−134.1

1p22

87012759

87046437

+

protein-coding

CA1

ENSG00000133742

−85.4

8q21

86239837

86291243

protein-coding

SLC4A4

ENSG00000080493

−60.8

4q21

71092756

71572087

+

protein-coding

AQP8

ENSG00000103375

−51.8

16p12

25227052

25240261

+

protein-coding

ZG16

ENSG00000174992

−45.6

16p11

29778240

29782973

+

protein-coding

MS4A12

ENSG00000071203

−43.8

11q12

60260251

60274903

+

protein-coding

CA2

ENSG00000104267

−37.2

8q21

85463852

85481493

+

protein-coding

GUCA2B

ENSG00000044012

−35.7

1p34

42619092

42621495

+

protein-coding

CHGA

ENSG00000100604

−35.2

14q32

93389425

93401638

+

protein-coding

GUCA2A

ENSG00000197273

−35.2

1p34

42628362

42630395

protein-coding

CLCA1

ENSG00000016490

−34.8

1p22

86934051

86965977

+

protein-coding

SLC26A3

ENSG00000091138

−33.7

7q31

107405912

107443678

protein-coding

UGT2B17

ENSG00000197888

−31

4q13

69402902

69434245

protein-coding

Down-regulated miRNA transcripts

hsa-miR-215

ENSG00000207590

−11

1q41

220117853

220117962

+

hsa-miR

hsa-miR-139-5p

ENSG00000272036

−8

11q13

72615063

72615130

hsa-miR

hsa-miR-133a

ENSG00000207764

−7.1

20q13

61162119

61162220

+

hsa-miR

hsa-miR-378c

ENSG00000264803

−7

10q26

130962588

130962668

hsa-miR

hsa-miR-378d

ENSG00000263631

−6.7

4p16

5923275

5923328

hsa-miR

hsa-miR-422a

ENSG00000199156

−6.6

15q22

63870930

63871019

hsa-miR

hsa-miR-375

ENSG00000198973

−6.4

2q35

219866362

219866431

hsa-miR

hsa-miR-378f

ENSG00000264926

−6.3

1p36

23929070

23,929,147

+

hsa-miR

hsa-miR-378i

ENSG00000263463

−6.1

22q13

41923222

41923297

hsa-miR

hsa-miR-378g

ENSG00000263526

−5.6

1p21

94745860

94745900

hsa-miR

hsa-miR-133b

ENSG00000199080

−5.4

6p22

52148923

52149041

+

hsa-miR

hsa-miR-138

ENSG00000207954

−4.5

3p21

44114212

44114310

+

hsa-miR

hsa-miR-378e

ENSG00000263831

−4.2

5q35

169455492

169455570

+

hsa-miR

hsa-miR-143

ENSG00000208035

−3.7

5q32

148808481

148808586

+

hsa-miR

q-values < .01; T: tumoral samples; Non-T: non-tumoral samples; hsa-miR: human micro-RNA.

Classification of sCRC tumors vs non-tumoral colorectal tissues based on the gene expression profile (GEP) of those transcripts more strongly deregulated in primary tumors and their liver metastases.

Figure 2: Classification of sCRC tumors vs non-tumoral colorectal tissues based on the gene expression profile (GEP) of those transcripts more strongly deregulated in primary tumors and their liver metastases. (Panel A) Biplot analysis of 23 primary colorectal tumors (orange triangles) vs. 10 non-tumoral colorectal tissues (blue circles) from our sCRC patient series. (Panel B) Biplot analysis of 47 primary colorectal tumors (orange triangles) vs. 25 non-tumoral colorectal tissue samples (blue circles) from an independent external validation dataset (GEO database, accession number GSE21510). (Panel C) Biplot analysis of 19 liver metastases (red squares) vs. 10 non-tumoral colorectal tissues (blue circles) from our sCRC patient series. (Panel D) Biplot analysis of 24 liver metastases (red squares) vs. 23 non-tumoral colorectal tissues (blue circles) from an independent external validation dataset (GEO database, accession number GSE35834).

Overall, liver metastases from the 19 colorectal cancer patients analyzed systematically showed deregulated transcripts of those genes identified as being also deregulated in their paired primary colorectal carcinomas (Table 2). Thus, both the primary tumors and their paired liver metastases showed overexpression of FOXQ1, MMP7, CLDN1 and TACSTD2 mRNAS and the miR-4417, miR-503, miR-1290, miR-3687, miR-183, miR-224 and miR-1246 miRNAs, together with down-regulated levels of the CLCA4, CA1, AQP8, ZG16, GUCA2B and SLC26A3 mRNAS and the miR-215, miR-133a, miR-375, miR-133b and miR-138 miRNAs. Interestingly, most of these genes with highly deregulated expression in both primary tumors and their corresponding liver metastases have been previously found to be altered/involved in colorectal cancer (e.g., FOXQ1, MMP7, TACSTD2 CLCA4, CA1, AQP8, ZG16, GUCA2B and SLC26A3) and/or they have been identified as genes that are relevant to the metastatic process (e.g., FOXQ1, MMP7, TACSTD2 CLCA4, CA1, AQP8 and SLC26A3); similarly the miRNAs, miR-503, miR-3687, miR-215, miR-133a, miR-375, miR-183, miR-1290, miR-224, miR-1246, have also been reported to be typically altered in CRC. Despite the similarities observed between the GEP of primary and metastatic tumors, some transcripts were found to be specifically deregulated in liver metastases while expressed at normal levels in their paired primary tumors, reflecting either an increased genomic instability of metastatic cells or their adaption to the liver microenvironment (Table 3). Newly deregulated metastatic transcripts included overexpression of APOA1, HRG, UGT2B4, RBP4 and ADH4 mRNAS and the miR-3180-3p, miR-3197, miR-3178, miR-4793 and miR-4440 miRNAs, together with decreased expression of the IGKV1-39, IGKC, IGKV1-27, FABP4 and MYLK mRNAS and the miR-363, miR-1, miR-143, miR-27b and miR-28-5p miRNAs. Furthermore, once the 19 paired liver metastasis and primary tumor samples were specifically compared (paired analysis) 52 mRNAS (14 down- and 38 up-regulated genes) and only two over-expressed miRNAs were observed (miR-122 and miR-4322; Supplementary Table 2), these including the most differentially expressed mRNAs and miRNAs identified with the unpaired comparison of all samples. Of note, among the regulated miRNAs in both the paired and unpaired comparisons was miR-122, a miRNA which is highly expressed in the liver, wich could potentially be due to the presence of normal liver tissue in the metastasis samples. In order to determine the degree of contamination by residual liver cells in metastatic tissues, we analyzed the expression levels of 41 liver-associated genes, as selected from the Tissue-specific Gene Expression and Regulation database (TIGER) plus miR-122 in normal liver (n = 5), CRC liver metastases (n = 19), normal colorectal mucosa (n=9) and primary CRC tumors (n = 23). All 41 liver-specific genes showed similar expression levels in CRC liver metastases, normal colorectal mucosa and primary CRC tumors, while they were highly expressed in the normal liver samples (Supplementary Figure 1). In contrast, miR122 was highly expressed in the liver and CRC liver metastases, whereas it showed lower expression levels in normal mucosa and primary CRC tumors, suggesting that over-expression of miR122 in liver metastatic tissues was not specifically due to residual liver cell-associated background in the metastatic CRC tissues.

Table 2: Top mRNAs and miRNAs up- and down-regulated in colorectal liver metastases (n = 19) vs. non-tumoral colorectal tissues (n = 10)

Gene Name

Gene ID

Fold Change T vs. Non-T

Chr. band

Start (bp)

Stop (bp)

Strand

Transcript description

Up-regulated mRNA transcripts

FGA

ENSG00000171560

157.6

4q28

154583126

154590766

protein-coding

ALB

ENSG00000163631

96.2

4q13

73397114

73421412

+

protein-coding

CRP

ENSG00000132693

84.6

1q23

159712289

159,714,589

protein-coding

HP

ENSG00000257017

75.9

16q22

72054566

72,061,056

+

protein-coding

FGB

ENSG00000171564

64.2

4q28

154562956

154572763

+

protein-coding

MMP7

ENSG00000137673

59.2

11q22

102520508

102530753

protein-coding

APOA2

ENSG00000158874

54.4

1q23

161222292

161223631

protein-coding

ORM1

ENSG00000228278

50

9q31

114323023

114326479

+

protein-coding

APOA1

ENSG00000118137

36.1

11q23

116835751

116837950

protein-coding

CLDN1

ENSG00000163347

34.2

3q28

190305701

190322475

protein-coding

FOXQ1

ENSG00000164379

31.2

6p25

1312440

1314758

+

protein-coding

TACSTD2

ENSG00000184292

29.2

1p32

58575423

58577773

protein-coding

Up-regulated miRNA transcripts

hsa-miR-122

ENSG00000207778

1318.7

18q21

58451068

58451176

+

hsa-miR

hsa-miR-4417

ENSG00000264341

19.9

1p36

5564071

5564143

+

hsa-miR

hsa-miR-1290

ENSG00000221662

16.1

1p34

18897071

18897148

hsa-miR

hsa-miR-3687

ENSG00000264063

9.7

21p11

9826203

9826263

+

hsa-miR

hsa-miR-503

ENSG00000208005

9

Xq26

133680358

133680428

hsa-miR

hsa-mir-885

ENSG00000216135

8.9

3p25

10394481

10394562

hsa-miR

hsa-miR-183

ENSG00000207691

8.6

7q32

129774905

129775014

hsa-miR

hsa-miR-224

ENSG00000207621

8.4

Xq28

151958578

151958658

hsa-miR

hsa-miR-1246

ENSG00000207584

8.1

2q31

176600980

176601052

hsa-miR

Down-regulated mRNA transcripts

CLCA4

ENSG00000016602

−216.3

1p22

87012759

87046437

+

protein-coding

CA1

ENSG00000133742

−142.4

8q21

86239837

86291243

protein-coding

MS4A12

ENSG00000071203

−81.7

11q12

60260251

60274903

+

protein-coding

ZG16

ENSG00000174992

−66.5

16p11

29778240

29782973

+

protein-coding

SLC26A3

ENSG00000091138

−65.1

7q31

107405912

107443678

protein-coding

CLCA1

ENSG00000016490

−65

1p22

86934051

86965977

+

protein-coding

ACTG2

ENSG00000163017

−61

2p13

73892314

73892314

+

protein-coding

AQP8

ENSG00000103375

−55

16p12

25227052

25240261

+

protein-coding

CA2

ENSG00000104267

−53.6

8q21

85463852

85481493

+

protein-coding

FCGBP

ENSG00000090920

−52

19q13

39863323

39934626

protein-coding

GUCA2B

ENSG00000044012

−43.8

1p34

42619092

42621495

+

protein-coding

Down-regulated miRNA transcripts

hsa-miR-133a

ENSG00000207764

−22.5

20q13

61162119

61162220

+

hsa-miR

hsa-miR-133b

ENSG00000199080

−10.8

6p12

52148923

52149041

+

hsa-miR

hsa-miR-10b

ENSG00000207744

−7.5

2q31

176150303

176150412

+

hsa-miR

hsa-miR-215

ENSG00000207590

−7

1q41

220117853

220117962

+

hsa-miR

hsa-miR-363

ENSG00000207572

−5.9

Xq26

134169361

134169473

hsa-miR

hsa-miR-143

ENSG00000208035

−5.4

5q32

148808481

148808586

+

hsa-miR

hsa-miR-497

ENSG00000273895

−5.2

17p13

7017911

7018022

hsa-miR

hsa-miR-1

ENSG00000174407

−5.2

20q13

62550453

62570764

+

hsa-miR

hsa-miR-138

ENSG00000207954

−4.9

3p21

44114212

44114310

+

hsa-miR

hsa-miR-375

ENSG00000198973

−4.5

2q35

219866362

219866431

hsa-miR

q-values < .01; T: tumoral samples; Non-T: non-tumoral samples; hsa-miR: human micro-RNA.

Table 3: Top up- and down-regulated mRNAs and miRNAs differentially expressed in colorectal liver metastases (n = 19) vs. non-tumoral colorectal tissues (n = 10) vs. primary tumors (n = 23)

Gene Name

Gene ID

Fold Change T vs. Non−T

Chr. band

Start (bp)

Stop (bp)

Strand

Transcript description

Up-regulated mRNA transcripts

APOA1

ENSG00000118137

10.2

11q23

116835751

116837950

protein-coding

HRG

ENSG00000113905

10.0

3q27

186660216

186678240

+

protein-coding

UGT2B4

ENSG00000156096

9.3

4q13

69480165

69526014

protein-coding

RBP4

ENSG00000138207

9.0

10q23

93591687

93601744

protein-coding

ADH4

ENSG00000198099

8.3

4q22

99123657

99157792

protein-coding

SAA2-SAA4

ENSG00000148965

8.1

11p15

18231355

18248674

protein-coding

ITIH1

ENSG00000055957

7.9

3p21

52777586

52792068

+

protein-coding

CFHR3

ENSG00000116785

7.6

1q32

196774795

196795406

+

protein-coding

HPX

ENSG00000110169

7.5

11p15

6431038

6442617

protein-coding

CFHR2

ENSG00000080910

7.1

1q31

196943759

196959226

+

protein-coding

PLG

ENSG00000122194

7.1

6q26

160702193

160754054

+

protein-coding

Up-regulated miRNA transcripts

hsa-miR-3180-3p

ENSG00000257563

4.8

16p12

18402178

18402271

hsa-miR

hsa-miR-3197

ENSG00000263681

4.7

21q22

41167557

41167629

+

hsa-miR

hsa-miR-3178

ENSG00000266232

4.2

16p13

2531922

2532005

hsa-miR

hsa-miR-4793

ENSG00000263898

4.1

3p21

48644194

48644280

hsa-miR

hsa-miR-4440

ENSG00000266109

4.0

2q37

239068817

239068914

hsa-miR

hsa-miR-4486

ENSG00000265210

3.6

11p15

19575310

19575372

+

hsa-miR

hsa-miR-150-star

ENSG00000207782

3.5

19q13

49500762

49500873

hsa-miR

hsa-miR-483-5p

ENSG00000207805

3.5

11p15

2134111

2134222

hsa-miR

hsa-miR-642b

NA

3.4

19q13

45674932

45675008

hsa-miR

hsa-miR-1247

ENSG00000277601

3.4

14q32

101560287

101560422

hsa-miR

Down-regulated mRNA transcripts

IGKV1-39

ENSG00000242371

−15.9

2p11

89319,625

89320146

protein-coding

IGKC

ENSG00000239975

−15.0

2p12

88857161

89085723

protein-coding

IGKV1-27

ENSG00000244575

−13.3

2p11

89213417

89,213,928

protein-coding

FABP4

ENSG00000170323

−13.1

8q21

81478419

81483263

protein-coding

MYLK

ENSG00000065534

−11.5

3q21

123610049

123884331

protein-coding

PLN

ENSG00000198523

−11.3

6q22

118548279

118560730

+

protein-coding

RBPMS2

ENSG00000166831

−11.0

15q22

64739892

64775587

protein-coding

CNN1

ENSG00000130176

−9.8

19p13

11538717

11550324

+

protein-coding

SPINK4

ENSG00000122711

−9.8

9p13

33218365

33248567

+

protein-coding

LMOD1

ENSG00000163431

−9.3

1q32

201896452

201946588

protein-coding

MYH11

ENSG00000133392

−9.3

16p13

15703135

15857033

protein-coding

Down-regulated miRNA transcripts

hsa-miR-363

ENSG00000207572

−5.9

Xq26

134169361

134169473

hsa-miR

hsa-miR-1

ENSG00000174407

−5.2

20q13

62550453

62570764

+

hsa-miR

hsa-miR-143

ENSG00000208035

−3.9

5q32

149428918

149429023

+

hsa-miR

hsa-miR-27b

ENSG00000207864

−2.1

9q22

95085435

95085542

+

hsa-miR

hsa-miR-28-5p

ENSG00000207651

−2.0

3q28

188688781

18868866

+

hsa-miR

hsa-miR-99b

ENSG00000207550

−1.7

19q13

51692612

51692681

+

hsa-miR

hsa-miR-152

ENSG00000207947

−1.7

17q21

48037154

48037262

hsa-miR

hsa-miR-125b

ENSG00000207971

−1.5

11q24

122099757

122099844

hsa-miR

hsa-miR-3064

ENSG00000265695

−1.5

17q23

64500773

64500839

hsa-miR

hsa-miR-30d

ENSG00000199153

−1.4

8q24

134804876

134804945

hsa-miR

q-values < .01; T: tumoral samples; Non-T: non-tumoral samples; hsa-miR: human micro-RNA.

Functional characterization of deregulated GEP in metastatic sCRC

Analysis of the biological and functional significance of the deregulated GEPs observed in our metastatic colorectal tumors, revealed 58 significantly altered canonical pathways in primary CRC tumors (vs. non-tumoral colorectal tissue) and 65 associated with liver mestastases (vs. non-tumoral colorectal tissue) (Figure 1). Those pathways specifically altered in primary tumors (Supplementary Table 3) were associated with an abnormally increased expression of genes that are involved in focal adhesion (e.g., PRKCA, CRK, and BCL2), PI3K-Akt signaling (e.g., PHLPP2, PRKCA, PPP2R3A and KIT) and cancer pathways (e.g., COL4A5, LAMC1 and BCL2L1). Conversely, canonical pathways found to be specifically deregulated in liver metastatic samples (Supplementary Table 4) included multiple genes related to intercellular adhesion and the metastatic processes (e.g., IGF1R, PIK3CA, PTEN and EGFR), endocytosis (e.g., the PDGFRA, SMAD2, ERBB3, PML and FGFR2), and the cell cycle (e.g., SMAD2, CCND2, E2F5 and MYC), as well as, the TGFβ signaling pathway (e.g., RHOA, SMAD2, SMAD4, SMAD5, SMAD6, BMPR1A, SMAD7 and MYC), with the FDR values significantly higher for the liver metastases vs. primary tumors (≥ 0.0005 to 2.075E-13).

Validation of tumor markers with high discriminating power between primary tumors and non-tumoral colorectal tissues

The discriminating power of the top mRNAs found to be deregulated (up-regulated and down-regulated) in common in primary sporadic colorectal tumors (n = 23) and colorectal liver metastases (n = 19) [vs. non-tumoral colorectal tissues (n = 10)] was further validated using GEP data from two independent series of sCRC available at the public GEO database (47 primary tumors vs. 25 non-tumoral colorectal tissues and 24 liver metastases vs. 23 non-tumoral colorectal tissues; Figure 2). In line with those findings described above for our sCRC cases, all primary tumors and non-tumoral samples from the two validation cohorts could be classified as having a sCRC-associated GEP based on overexpression of FOXQ1, MMP7, CLDN1, TACSTD2, COL11A1, KIAA1199, CTHRC1, DUSP27, KRT23, SRPX2 and NFE2L3 and decreased expression of CLCA4, CA1, SLC4A4, AQP8, ZG16, MS4A12, CA2, GUCA2B, CHGA, GUCA2A, CLCA1, SLC26A3 and UGT2B17, respectively. Similarly, 23/24 (96%) liver metastatic samples were also properly classified as distinct from non-tumoral colorectal tissue based on overexpression of APOA1, HRG, UGT2B4, RBP4, ADH4, SAA2-SAA4, ITIH1, CFHR3, HPX, CFHR2 and PLG, while all non-tumoral samples showed decreased expression of IGK1-39, IGKC, IGKV1-27, FABP4, MYLK, PLN, RBPMS2, CNN1, SPINK4, LMOD1 and MYH11 (Figure 2).

miRNAs genes that potentially regulate gene expression in metastatic sCRC

In order to determine the impact of the miRNAs gene expression signature on the GEP of sCRC tumors, both the miRNA and mRNA gene expression data sets were combined to investigate potential correlations between miRNAs and mRNA genes which are specifically altered in colorectal liver metastases. Evaluation of each pair of potential miRNA-mRNA interacting genes identified potential interactions for 38 inversely correlated and 437 directly correlated (absolute R2 value ≥ 0.80; p < .0001) pairs of miRNA-mRNA genes. Based on currently available miRNA target prediction algorithms and databases, such interactions corresponded to only three predictable and 4 experimentally validated interactions for the inversely correlated pairs of miRNA-mRNA (Table 4). Of note, both the 4 experimentally validated pairs of mRNA/miRNA genes (miR-20a/DNAJB4, miR-497/PHF19, miR-335/PTPRH and miR-195/SLC23A3) and the 3 predicted miRNA-mRNA pairs (miR-363/LRRC1, miR-3124/PRRC2C and miR-363/ MIER3) were systematically altered in all 19 liver metastatic samples analyzed in this study.

Table 4: miRNA-mRNA interactions found in metastatic colorectal cancer by pearson correlation analysis of the expression signal identified for those transcripts differentially and exclusively (vs. primary tumors) expressed in colorectal liver metastases (n = 19) as detected by the affymetrix pimeview human gene expression array and the microRNA 3.0 expression array

miRNA

mRNA Gene Name

Gene ID

R2

Classification of Interaction

Source of validation/prediction

hsa-miR-20a

DNAJB4

ENSG00000162616

−0.84

Validated

miRTarBase

hsa-miR-497

PHF19

ENSG00000119403

−0.83

Validated

miRTarBase

hsa-miR-335

PTPRH

ENSG00000080031

−0.82

Validated

miRTarBase, DIANAmT

hsa-miR-195

SLC23A3

ENSG00000213901

−0.82

Validated

miRTarBase

hsa-miR-363

LRRC1

ENSG00000137269

−0.89

Predicted

MiRanda

hsa-miR-3124

PRRC2C

ENSG00000117523

−0.86

Predicted

Targetscan

hsa-miR-363

MIER3

ENSG00000155545

−0.81

Predicted

Targetscan

hsa-miR-10b-star

NBPF1

ENSG00000219481

−0.85

Not known

hsa-miR-30a

KIF4A

ENSG00000090889

−0.85

Not known

hsa-miR-31

QPRT

ENSG00000103485

−0.85

Not known

hsa-miR-1910

RASL12

ENSG00000103710

−0.84

Not known

hsa-miR-30a

SKA1

ENSG00000154839

−0.84

Not known

hsa-miR-195

SKA1

ENSG00000154839

−0.83

Not known

hsa-miR-3162

TFCP2L1

ENSG00000115112

−0.83

Not known

hsa-miR-486

SPC24

ENSG00000161888

−0.83

Not known

hsa-miR-497

SKA1

ENSG00000154839

−0.83

Not known

hsa-miR-885

RNF43

ENSG00000108375

−0.83

Not known

hsa-miR-195

F2RL1

ENSG00000164251

−0.82

Not known

hsa-miR-195

KIF4A

ENSG00000090889

−0.82

Not known

hsa-miR-30a

BORA

ENSG00000136122

−0.82

Not known

hsa-miR-4484

CASP1

ENSG00000137752

−0.82

Not known

hsa-miR-497

HMMR

ENSG00000072571

−0.82

Not known

hsa-miR-497

KIF4A

ENSG00000090889

−0.82

Not known

hsa-miR-497

PTTG1

ENSG00000164611

−0.82

Not known

hsa-miR-195

CKMT1A

ENSG00000223572

−0.81

Not known

hsa-miR-20a

PCSK5

ENSG00000099139

−0.81

Not known

hsa-miR-223

PDE9A

ENSG00000160191

−0.81

Not known

hsa-miR-363

MRPS17

ENSG00000239789

−0.81

Not known

hsa-miR-4443

RRP1

ENSG00000160214

−0.81

Not known

hsa-miR-150

TFCP2L1

ENSG00000115112

−0.80

Not known

hsa-miR-20a

RNF152

ENSG00000176641

−0.80

Not known

hsa-miR-20a

SOCS2

ENSG00000120833

−0.80

Not known

hsa-miR-30a

MS4A8

ENSG00000166959

−0.80

Not known

hsa-miR-363

CNBP

ENSG00000169714

−0.80

Not known

hsa-miR-363

UQCRC2

ENSG00000140740

−0.80

Not known

hsa-miR-497

CKMT1A

ENSG00000223572

−0.80

Not known

hsa-miR-497

F2RL1

ENSG00000164251

−0.80

Not known

hsa-miR-497

HDHD2

ENSG00000167220

−0.80

Not known

hsa.miR.378g

PMEPA1

ENSG00000124225

−0.80

Not known

p-values <.0001; R2: Pearson correlation coefficient.

DISCUSSION

In the present study we investigated the expression of both coding mRNA and non-coding RNA genes -including miRNA, small nucleolar and large intergenic RNAs- in paired primary tumor and liver metastases samples from primary colorectal carcinoma patients. To the best of our knowledge, this is the first study in which the overall coding and non-coding GEP of sCRC liver metastases are compared with their paired primary tumors and non-tumoral colorectal tissues, by high-resolution arrays. Overall, primary tumors and their paired metastases frequently revealed many genes to be altered in common at both sites; these findings support the existence of a very close genomic relationship between the GEP of primary colorectal tumors and their paired liver metastases, as it has also been suggested to occur at the DNA level [16]. Deregulated expression observed in common in both groups of (paired) tumor samples included increased expression of genes linked to liver metastatic sCRC, tumor development, growth (TACSTD2) [17] and invasiveness (FOXQ1, MMP7 and CLDN1) [18, 19], together with decreased expression of genes related to differentiation (CLCA4, CLCA1 and AQP8) [20, 21], apoptosis (CA1) [22], immunity (ZG16) [23], and protection (FCGBP and CA2) [24] of CRC cells. Thus, the functional networks most strongly affected in primary tumors included those involved in focal adhesion, PI3K-Akt and ErbB signaling and other cancer pathways, FcδR-mediated phagocytosis, endocytosis, bacterial invasion of epithelial cells, plus the TP53 signaling pathway, all of which have been previously shown to be directly involved in the development and progression of sCRC [2529].

Another functional gene network which was also frequently altered in sCRC liver metastases was the TGFβ signaling pathway, a network that plays a critical role in the regulation of cell growth, differentiation and development in a broad range of neoplasms [7]. In fact, such findings have led to the speculation that TGFβ signaling might be responsible, at least in part, for the aggressiveness of CRC tumors in our series, through an increased invasive capacity which is a prerequisite for the settlement and growth of distant metastasis [18]. In this regard, recent studies in which the miRNAS signature of metastatic CRC has been investigated, have also identified a network of regulatory miRNA-driven interactions that regulate expression of key genes associated with TGFβ signaling and metastasis, including the miR-378 gene family [30]. In line with these observations, here we found the miRNA-378c, -378d, -378f, -378i, -378g and -378e to be among the most strongly down-regulated miRNAs in primary CRC tumors. Zhang et al. [30], evaluated the levels of miR-378 in CRC cell lines and 84 paired CRC cancer and normal adjacent mucosa tissues, their results highlighting miR-378 expression to be an independent prognostic factor potentially due to its ability to inhibit cell growth and invasion in CRC. Most interestingly, here we found that miR-378g could directly target PMEPA1, a gene that plays a relevant role in the TGF-β signaling pathway [31, 32]. In this regard, it has been recently shown that the TGF-β signaling regulator PMEPA1 suppresses prostate cancer metastases to bone [32]. Despite this, the impact of an altered PMEPA1 expression on CRC, as well as the potential underlying mechanisms for such association, still remain to be investigated. Moreover, several miRNA whose expression was strongly down-regulated in metastatic CRC samples, are also involved in the TGFβ signaling pathway; these included the miR-133a, miR-133b, miR-1 and miR-375 miRNAs [33].

Previous studies in which the miRNA signature of metastatic CRC has been investigated, have identified a unique pattern of miRNA which are differentially expressed in metastatic tumors and that include a network of regulatory miRNA-driven interactions involved in the expression of key genes associated with the epithelial to mesenchymal transition (EMT) and tumor metastasis, such as the miR-200 gene family [15]. However, in these studies the transcriptional profiles of the metastatic liver was defined though comparison of the gene expression profile of CRC metastatic tissues against normal liver, despite the background GEP of the tumor present in the liver derives from cells of the (tumoral) intestinal mucosa and not from liver tissues. Similarly, Lin et al. [34] using supervised SAM analysis, identified 963 unique genes to be significantly overexpressed in colorectal liver metastases vs. primary CRC (and normal liver tissue). However, several of the differently expressed genes are known to be up-regulated in normal liver vs colon tissues (i.e.: ARG1, BAAT, BHMT, CDO1, CRHBP, F11, F5, FGL1, HAMP, HAMP and ITIH2).

Of note, miR-122 emerged as the most up-regulated miRNA gene in liver metastases vs. both non-tumoral colorectal mucosa and primary CRC tissues. Overexpression of miR-122 and the simultaneus suppression of its target gene, cationic amino acid transporter 1, has been shown to be involved in the development of colorectal liver metastasis [35]. However, Pizzini et al. [36] have previously suggest that overexpression of miR-122 in CRC liver metastatic tissues could be due to the presence of normal residual liver tissue. Based on an extensive analysis of other liver associated genes, our data suggests this might not be the case and that miR-122 could in fact the overexpression by metastatic liver cells. In this regard, it should be noted that an interesting cross-talk has been reported between hepatocarcinoma cells, through which cells expressing miR-122 send this miRNA via microvesicles to inhibit the proliferation of miR-122 deficient cells. In a reciprocal process, miR-122 deficient cells secrete insulin-like growth factor to decrease miR-122 expression in miR-122 expressing cells [37].

Our results also showed miR-10b to be down-regulated in liver metastatic tissues vs non-tumoral colorectal mucosa. The expression levels of this miRNA in metastataic samples (vs. primary tumors) have previously been correlated with CRC patients survival [36]. In this regard, Hur et al have recently identified a metastasis-specific microRNA signature that includes high miR-10b expression in primary tumors, to be an independent predictor of distant metastasis [38]. In addition, it is also well known that miR-10b functionally contributes to tumor invasion and metastasis in breast cancer [39], and to a worse prognosis in both esophageal [40] and pancreatic cancer patients [41]. Despite all the above, delineation of the specific functional effects of miRNA levels on the expression of mRNA transcripts still remains a challenge. Here we identified several miRNAs whose expression levels were significantly correlated with the amount of mRNA of specific genes. Among other pairs of miRNA-mRNA genes identified, the miR-20a and the DNAJB4 gene transcript emerged in our series, as significantly correlated. DNAJB4 (DnaJ heat shock protein family member B4) is a chaperone with a strong tumor suppressor effect in CRC whose down-regulation has been associated with patient outcome [42]. In this regard, miR-20a has also been shown to induce EMT, to regulate the migration and invasion of SW480 cells [43] and to contribute to an increased chemoresistance of CRC [44]. Of note, here we did not find any (inverse) correlation between the levels of expression of miR-20a and the mRNA levels of other genes reported to be altered IN parallel in CRC such as the PCSK5, RNF152 and SOCS2 genes [4547]. Other miRNAs which have been found to be altered in CRC include the miR-497, which might mediate overexpression of genes involved in cell proliferation (e.g. PHF19, SKA1, KIF4A) and genes associated with cell migration and invasion (e.g. PTTG1, F2RL1) as well as genes related to the transfer of high energy phosphate from mitochondria, potentially related to the failure to eliminate cancer cells via apoptosis (e.g. CKMT1A) [48, 49].

Interestingly, clearcut discrimination between primary CRC, liver metastases and non-tumoral colorectal tissue could be obtained via those mRNAs found to be most strongly up- and down-regulated exclusively in primary tumors and their liver metastases, as also confirmed in two external series of 47 metastatic CRC [50] and 24 colorectal liver metastases [36]. These results indicate that these gene signatures could potentially serve in the future as prior knowledge for the discovery of biomarker candidates for primary sCRC tumors for early diagnosis, more efficient treatment and/or monitoring of metastatic CRC. In line with this hypothesis, strong expression of the MMP7, CLDN1, TACSTD2, CTHRC1, KRT23 and SRPX2 genes has been previously reported at the protein level in CRC tissues, their expression been also associated with tumor progression and an increased angiogenesis [51]; of note most of these proteins (i.e.: MMP7, TACSTD2, CTHRC1 and KRT23) have also been found to be secreted and to be present in both tumor tissues and the plasma from CRC patients [52]. Therefore, secretion of these proteins outside the tumor cell supports the potential utility of these genes as serum biomarkers for early diagnosis and monitoring of CRC patients. However, additional studies are still required to further validate in non-metastatic CRC the mRNA and miRNA expression profiles here described.

In summary, here we report on a high number of a mRNA and miRNA that are altered in common in (paired) primary and metastatic colorectal tumors, and that allow clearcut distinction between the tumoral and normal colon tissues based on a few mRNAs potentially critical in either the metastatic process and/or the adaption of metastatic cells to the metastatic niche/environment. In this regard, activation of genes associated with the TGFβ signaling pathway emerged as potentially relevant candidate genes in CRC metastasis.

MATERIALS AND METHODS

Patients and samples

Tissue specimens from 23 sporadic colorectal adenocarcinomas and 19 paired liver metastases (n = 42 samples) were obtained from 23 patients with metastatic lesions susceptible of being resected (16 males and 7 females; median age of 66 years, ranging from 48 to 80 years) after informed consent had been given by each subject; in four patients no metastatic liver tissue was available. The studied cases corresponded to 23 consecutive metastatic colorectal cancer patients. Patients underwent surgical resection of both primary and metastatic tumor tissues at the Department of Surgery of the University Hospital of Salamanca (Salamanca, Spain), prior to any cytotoxic therapy was given. Tumor diagnosis and classification was performed according to the AJCC criteria [53]. According to tumor grade, 11 cases were classified as well-differentiated tumors, 10 as moderately- and 2 as poorly-differentiated carcinomas. In all cases, histopathological grade was systematically confirmed in a second independent evaluation by an experienced pathologist (M.A.A and OB). Median follow-up at the moment of closing the study was of 25 months (range: 11–39 months).

Primary tumors were localized in the rectum (n = 11) and either in the right (cecum, ascending or transverse) or the left (descending and sigmoid) colon (n = 12). The mean size of the primary tumors was of 5.4 ± 2.0 cm with the following distribution according to their TNM stage at diagnosis [54]: T3N0M0, 2 cases; T3N1M1, 8; T3N1M0, 1; T3N0M1, 3; T3N2M1, 2; T4N0M1, 2; T4N1M1, 1; T4N2M1, 2; T2N0M0, 1 and; T3N2M0, 1 case. Eighteen liver metastases were synchronous and 5 were metachronous metastases. The mean size of liver metastases was of 3.6 ± 2.1 cm. The most relevant clinical and laboratory data for each individual metastatic colorectal cancer patient studied is summarized in Supplementary Table 1.

Colorectal tissue samples not required for diagnostic purpose were collected immediately after surgical resection, snap frozen and stored in OCT at −80°C (Tumor Biobank of the University Hospital of Salamanca, Red de Bancos de Tumores de Castilla y León, Salamanca, Spain). Once the histopathological diagnosis had been established, sections from paraffin-embedded tissue samples were cut from three different areas representative of the tumor tissue with > 70% tumor cell infiltration by hematoxylin-eosin staining, after excluding stroma-enriched tumor areas. In order to enrich the tumor cells, the neighbour areas of those containing ≥ 70% tumor cells, as well as liver tissue samples were then microdissected from the frozen tumor tissue samples stored in OCT, following the pathologist criteria. Normal mucosa samples (n = 9) were taken at a minimum distance of 10 cm from the tumor site. The study was approved by the local ethics committee (University Hospital of Salamanca, Salamanca, Spain).

RNA extraction and gene expression profiling (GEP) microarray studies

For GEP, sample preparation was performed as described in the Affymetrix GeneChip Expression Analysis Manual (Santa Clara, CA, USA). Briefly, each frozen tissue (≥ 0.3 g) was crushed to powder at cryogenic temperatures and homogeneized in Trizol (Life Technologies Inc, Rockville, MD). Total RNA was then extracted using the miRNeasy mini kit according to the instructions of the manufacturer (Qiagen, Valencia, CA); subsequently, the quality and integrity of the RNA was evaluated in an Agilent 2100 Bioanalyzer (Agilent Technologies Inc, Santa Clara, CA). Total RNA (100–1000 ng) from both tumoral and non-tumoral colorectal tissues was hybridized to both the Affymetrix PimeView Human Gene Expression and the microRNA 3.0 Expression arrays, following the instructions of the manufacturer. Fluorescence signals were detected using the GeneChip Scanner 3000 7G (Affymetrix) and data stored as .CEL files. GEP raw data has been deposited in the Gene Expression Omnibus (GEO) database with the GSE81582 accession number, includes data based on sets of 49,395 and 5,683 probes for the Affymetrix PimeView Human Gene Expression microarray and the microRNA 3.0 microarray, respectively.

For data analysis, GEP raw data was normalized with the Robust Multi-array Average (RMA) algorithm, which included sequential background correction, intra- and inter-microarray well normalization, probe set summarization and calculation of expression signals [55]. Unsupervised classification of samples and genes was performed by MultiDimensional Scaling (MDS) and hierarchical clustering analyses (HCA) based on the expression signal detected for each gene for each probe set, and the Simfit statistical software (http://www.simfit.org.uk/). Clustering was run using Euclidean distances and the linkage method group average. Differentially expressed (miRNA and mRNA) genes between tumoral and non-tumoral samples were identified by supervised two-class unpaired Significance Analysis of Microarray (SAM) [56] based on a q-value cut off ≤ .01 and an absolute fold change cutoff ≥ 2.0. Differentially expressed mRNA and miRNA in pairwise group comparisons, involving groups of samples paired per patient (n = 19 paired liver metastasis vs 19 primary tumor samples) were calculated with SAM using a two-class pair wise design. The false discovery rate (FDR) cutoff for statistical significance was set at values < .01.

For the identification of miRNA candidates acting as gene-regulators in colorectal samples, Pearson correlation analysis was performed to identify significant associations between deregulated miRNA and mRNA gene transcripts in both primary colorectal tumors (vs. non-tumoral colorectal tissues) and colorectal liver metastases (vs. non-tumoral colorectal tissues) through the psych R-package, based on an adjusted FDR of ≤ .05. Each potential miRNA-mRNA interaction identified was subsequently evaluated against databases of experimentally validated miRNA interactions (TarBase 6.0 and miRWalk-database) and miRNA target prediction tools (DIANA-microT-CDS v5.0, miRWalk-database and miRecords) [57, 58]. Functional enrichment analysis of deregulated genes, as well as analysis of canonical pathways, was based on the use of the WebGestalt suite (http://bioinfo.vanderbilt.edu/webgestalt).

Enrichment of kyoto encyclopedia of genes and genomes (KEGG) pathways

To identify significantly altered KEGG pathways for each comparison performed, we applied the hypergeometric distribution statistical test was applied to calculate the probability of overlap of the deregulated transcripts, targets of significantly differentially expressed miRNAs, and gene expression involved in KEGG pathways; p-values were corrected using the Benjamini-Hochberg .

Validation of miRNA expression profiles by quantitative real-time PCR (QR-PCR)

TaqMan miRNA expression assays were used to validate GEP in the same samples that were used for the microarray studies, via the Step One Plus Real-Time PCR System -Applied Biosystems (ABI), Foster City, CA, USA- according to the manufacturer’s instructions. The assays ID for the miRNAs studied were as follows: 002245 (hsa-miR-122), 2246 (hsa-miR-133a), 002249 (hsa-miR-143) and 461910 (hsa-miR-4417). Each PCR was carried out in duplicate in a final volume of 10 uL using the TaqMan Fast Universal Mastermix (ABI) and the following cycling parameters: incubation at 95°C (20s), followed by 50 cycles at 95°C (1s) and an incubation at 60°C (20s). miRNA expression data was normalized against the RNU43 internal control, and it was further analyzed using the StepOne software (v2.0; ABI). The relative amounts of the quantified miRNAs were calculated using the following equation: 2−ΔCT (ΔCT = CT GENE-CT RNU43) expressed as arbitrary units (AU). For all genes evaluated, RQ-PCR results showed a high degree of correlation with microarray data (r2 ≥ 0.79, p < .0001; Supplementary Figure 2).

External validation series of sCRC tumors

External validation of the predictive value of those differentially expressed genes found in our series to discriminate between primary tumors and non-tumoral colorectal tissues, was performed in a group of previously reported metastatic sCRC patients (n = 47) from whom GEP array data files (Affymetrix Human Genome U133 Plus 2.0 Array) are publicly available at the GEO database (accession number GSE21510) [50]. Additionally, to discriminate between liver metastases and non-tumoral colorectal tissues, we performed an external validation in another group of previously reported metastatic sCRC patients (n = 24) from whom GEP array data files (Affymetrix Human Exon 1.0 ST Array) are also publicly available at the GEO database (accession number GSE35834) [36].

Downloaded data CEL files were normalized using the RMA algorithm and overlapping probe sets were defined on the basis of probe specificity, using the GATExplorer server [59]. Probe sets with the best specificity to the interrogated genes were selected, and the expression signals detected for each gene for each probe set were further analyzed using the column metric preserving biplot assay [60] implemented in the SIMFIT statistical software (http://www.simfit.org.uk/).

External validation series of DEFB1, COL12A1 and PTGER3 gene expression

External validation of those genes found to be differentially expressed in our series between primary tumors and their corresponding liver metastases (DEFB1, COL12A1 and PTGER3), was performed in a group of previously reported metastatic sCRC patients (n = 18) from whom RNA-seq data files (Illumina, CA) are publicly available at the GEO database (accession number GSE50760) [61]; in line with our observations, these results confirmed the existence of significant differences in the expression of the DEFB1, COL12A1 and PTGER3 genes between paired primary tumor and their corresponding liver metastases (Supplementary Figure 3).

Other statistical methods

The Mann-Whitney U test and a linear regression model were used to evaluate the statistical significance of differences observed between groups and to explore the degree of correlation between different variables, respectively (SPSS version 15.0; IBM; NY; USA). P-values ≤ .05 were considered to be associated with statistical significance.

CONFLICTS OF INTEREST

None.

FUNDING

This work has been partially supported by grants from the Instituto de Salud Carlos III (ISCIII; Ministerio de Sanidad y Consumo, Madrid, Spain) (PI12/02053-FIS), Gerencia Regional de Salud de Castilla y León, Valladolid, Spain (GRS1302/A/16), Consejería de Sanidad (Junta de Castilla y Leon, Valladolid, Spain) (BIO/SA02/13 and BIO/SA46/14), RTICC from the ISCIII (RD12/0020/0035-FEDER, RD12/0036/0048-FEDER), Fundación Memoria de Don Samuel Solórzano Barruso, (Salamanca, Spain) and Fundación Eugenio Rodríguez Pascual, (Madrid, Spain). JM Sayagués and ME Sarasquete are supported by grants (CES11/004 and CP13/00080; respectively) from the ISCIII, Ministerio de Ciencia e Innovación, Madrid, Spain.

REFERENCES

1. Tsai HL, Lu CY, Hsieh JS, Wu DC, Jan CM, Chai CY, Chu KS, Chan HM, Wang JY. The prognostic significance of total lymph node harvest in patients with T2–4N0M0 colorectal cancer. J Gastrointest Surg. 2007; 11:660–5.

2. Macartney-Coxson DP, Hood KA, Shi HJ, Ward T, Wiles A, O’Connor R, Hall DA, Lea RA, Royds JA, Stubbs RS, Rooker S. Metastatic susceptibility locus, an 8p hot-spot for tumour progression disrupted in colorectal liver metastases: 13 candidate genes examined at the DNA, mRNA and protein level. BMC Cancer. 2008; 8:187.

3. Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, Nakamura Y, White R, Smits AM, Bos JL. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988; 319:525–32.

4. Fearon ER, Hamilton SR, Vogelstein B. Clonal analysis of human colorectal tumors. Science. 1987; 238:193–7.

5. Negri FV, Bozzetti C, Lagrasta CA, Crafa P, Bonasoni MP, Camisa R, Pedrazzi G, Ardizzoni A. PTEN status in advanced colorectal cancer treated with cetuximab. Br J Cancer. 2010; 102:162–4.

6. Lan YT, Jen-Kou L, Lin CH, Yang SH, Lin CC, Wang HS, Chen WS, Lin TC, Jiang JK, Chang SC. Mutations in the RAS and PI3K pathways are associated with metastatic location in colorectal cancers. J Surg Oncol. 2015; 111:905–10.

7. Xu X, Chen R, Li Z, Huang N, Wu X, Li S, Li Y, Wu S. MicroRNA-490–3p inhibits colorectal cancer metastasis by targeting TGFbetaR1. BMC Cancer. 2015; 15:1023.

8. Mollevi DG, Serrano T, Ginesta MM, Valls J, Torras J, Navarro M, Ramos E, Germà JR, Jaurrieta E, Moreno V, Figueras J, Capellà G, Villanueva A. Mutations in TP53 are a prognostic factor in colorectal hepatic metastases undergoing surgical resection. Carcinogenesis. 2007; 28:1241–6.

9. Mlcochova J, Faltejskova-Vychytilova P, Ferracin M, Zagatti B, Radova L, Svoboda M, Nemecek R, John S, Kiss I, Vyzula R, Negrini M, Slaby O1. MicroRNA expression profiling identifies miR-31–5p/3p as associated with time to progression in wild-type RAS metastatic colorectal cancer treated with cetuximab. Oncotarget. 2015; 6:38695–704. doi: 10.18632/oncotarget.5735.

10. Chang SW, Yue J, Wang BC, Zhang XL. miR-503 inhibits cell proliferation and induces apoptosis in colorectal cancer cells by targeting E2F3. Int J Clin Exp Pathol. 2015; 8:12853–60.

11. Zheng K, Liu W, Liu Y, Jiang C, Qian Q. MicroRNA-133a suppresses colorectal cancer cell invasion by targeting Fascin1. Oncol Lett. 2015; 9:869–74.

12. Gutierrez ML, Corchete L, Teodosio C, Sarasquete ME, del Mar AM, Iglesias M, Esteban C, Sayagues JM, Orfao A, Muñoz-Bellvis L. Identification and characterization of the gene expression profiles for protein coding and non-coding RNAs of pancreatic ductal adenocarcinomas. Oncotarget. 2015; 6:19070–86. doi: 10.18632/oncotarget.4233.

13. Kopetz S, Tabernero J, Rosenberg R, Jiang ZQ, Moreno V, Bachleitner-Hofmann T, Lanza G, Stork-Sloots L, Maru D, Simon I, Capellà G, Salazar R. Genomic classifier ColoPrint predicts recurrence in stage II colorectal cancer patients more accurately than clinical factors. Oncologist. 2015; 20:127–33.

14. Kelley RK, Van Bebber SL, Phillips KA, Venook AP. Personalized medicine and oncology practice guidelines: a case study of contemporary biomarkers in colorectal cancer. J Natl Compr Canc Netw. 2011; 9:13–25.

15. Mudduluru G, Abba M, Batliner J, Patil N, Scharp M, Lunavat TR, Leupold JH, Oleksiuk O, Juraeva D, Thiele W, Rothley M, Benner A, Ben-Neriah Y, et al. A Systematic Approach to Defining the microRNA Landscape in Metastasis. Cancer Res. 2015; 75:3010–9.

16. Munoz-Bellvis L, Fontanillo C, Gonzalez-Gonzalez M, Garcia E, Iglesias M, Esteban C, Gutierrez ML, Abad MM, Bengoechea O, De Las Rivas J, Orfao A, Sayagués JM. Unique genetic profile of sporadic colorectal cancer liver metastasis versus primary tumors as defined by high-density single-nucleotide polymorphism arrays. Mod Pathol. 2012; 25:590–601.

17. Furi I, Kalmar A, Wichmann B, Spisak S, Scholler A, Bartak B, Tulassay Z, Molnár B. Cell Free DNA of Tumor Origin Induces a ‘Metastatic’ Expression Profile in HT-29 Cancer Cell Line. PLoS ONE. 2015; 10:e0131699.

18. Peng X, Luo Z, Kang Q, Deng D, Wang Q, Peng H, Wang S, Wei Z. FOXQ1 mediates the crosstalk between TGF-beta and Wnt signaling pathways in the progression of colorectal cancer. Cancer Biol Ther. 2015; 16:1099–109.

19. Kinugasa T, Akagi Y, Ochi T, Tanaka N, Kawahara A, Ishibashi Y, Gotanda Y, Yamaguchi K, Shiratuchi I, Oka Y, Kage M, Shirouzu K. Increased claudin-1 protein expression in hepatic metastatic lesions of colorectal cancer. Anticancer Res. 2012; 32:2309–14.

20. Yang B, Cao L, Liu B, McCaig CD, Pu J. The transition from proliferation to differentiation in colorectal cancer is regulated by the calcium activated chloride channel A1. PLoS ONE. 2013; 8:e60861.

21. Zhu J, Li C, Ji W. Identification of genes in ulcerative colitis associated colorectal cancer based on centrality analysis of co-expression network. Neoplasma. 2015; 62:756–64.

22. Peng Y, Li X, Wu M, Yang J, Liu M, Zhang W, Xiang B, Wang X, Li X, Li G, Shen S. New prognosis biomarkers identified by dynamic proteomic analysis of colorectal cancer. Mol Biosyst. 2012; 8:3077–88.

23. Wang L, Hahnloser D, Boardman LA, Burgart LJ, French AJ, Thibodeau S. Loss of expression of zymogen granule protein 16 in colorectal cancer. Cancer Res Abstract. 2005; 1:452.

24. Yasui Y, Tanaka T. Protein expression analysis of inflammation-related colon carcinogenesis. J Carcinog. 2009; 8:10.

25. Pandurangan AK. Potential targets for prevention of colorectal cancer: a focus on PI3K/Akt/mTOR and Wnt pathways. Asian Pac J Cancer Prev. 2013; 14:2201–5.

26. Jung HC, Eckmann L, Yang SK, Panja A, Fierer J, Morzycka-Wroblewska E, Kagnoff MF. A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest. 1995; 95:55–65.

27. Marszalowicz GP, Snook AE, Magee MS, Merlino D, Berman-Booty LD, Waldman SA. GUCY2C lysosomotropic endocytosis delivers immunotoxin therapy to metastatic colorectal cancer. Oncotarget. 2014; 5:9460–71. doi: 10.18632/oncotarget.2455.

28. Szmida E, Karpinski P, Leszczynski P, Sedziak T, Kielan W, Ostasiewicz P, Sasiadek MM. Aberrant methylation of ERBB pathway genes in sporadic colorectal cancer. J Appl Genet. 2015; 56:185–92.

29. Caro XJ. New concepts in primary fibrositis syndrome. Compr Ther. 1989; 15:14–22.

30. Zhang GJ, Zhou H, Xiao HX, Li Y, Zhou T. MiR-378 is an independent prognostic factor and inhibits cell growth and invasion in colorectal cancer. BMC Cancer. 2014; 14:109.

31. Brunschwig EB, Wilson K, Mack D, Dawson D, Lawrence E, Willson JK, Lu S, Nosrati A, Rerko RM, Swinler S, Beard L, Lutterbaugh JD, et al. PMEPA1, a transforming growth factor-beta-induced marker of terminal colonocyte differentiation whose expression is maintained in primary and metastatic colon cancer. Cancer Res. 2003; 63:1568–75.

32. Fournier PG, Juarez P, Jiang G, Clines GA, Niewolna M, Kim HS, Walton HW, Peng XH, Liu Y, Mohammad KS, Wells CD, Chirgwin JM, Guise TA. The TGF-beta Signaling Regulator PMEPA1 Suppresses Prostate Cancer Metastases to Bone. Cancer Cell. 2015; 27:809–21.

33. Butz H, Racz K, Hunyady L, Patocs A. Crosstalk between TGF-beta signaling and the microRNA machinery. Trends Pharmacol Sci. 2012; 33:382–93.

34. Lin AY, Chua MS, Choi YL, Yeh W, Kim YH, Azzi R, Adams GA, Sainani K, van de Rijn M, So SK, Pollack JR. Comparative profiling of primary colorectal carcinomas and liver metastases identifies LEF1 as a prognostic biomarker. PLoS ONE. 2011; 6:e16636.

35. Iino I, Kikuchi H, Miyazaki S, Hiramatsu Y, Ohta M, Kamiya K, Kusama Y, Baba S, Setou M, Konno H. Effect of miR-122 and its target gene cationic amino acid transporter 1 on colorectal liver metastasis. Cancer Sci. 2013; 104:624–30.

36. Pizzini S, Bisognin A, Mandruzzato S, Biasiolo M, Facciolli A, Perilli L, Rossi E, Esposito G, Rugge M, Pilati P, Mocellin S, Nitti D, Bortoluzzi S, et al. Impact of microRNAs on regulatory networks and pathways in human colorectal carcinogenesis and development of metastasis. BMC Genomics. 2013; 14:589.

37. Basu S, Bhattacharyya SN. Insulin-like growth factor-1 prevents miR-122 production in neighbouring cells to curtail its intercellular transfer to ensure proliferation of human hepatoma cells. Nucleic Acids Res. 2014; 42:7170–85.

38. Hur K, Toiyama Y, Schetter AJ, Okugawa Y, Harris CC, Boland CR, Goel A. Identification of a metastasis-specific MicroRNA signature in human colorectal cancer. J Natl Cancer Inst. 2015; 107.

39. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007; 449:682–8.

40. Tian Y, Luo A, Cai Y, Su Q, Ding F, Chen H, Liu Z. MicroRNA-10b promotes migration and invasion through KLF4 in human esophageal cancer cell lines. J Biol Chem. 2010; 285:7986–94.

41. Nakata K, Ohuchida K, Mizumoto K, Kayashima T, Ikenaga N, Sakai H, Lin C, Fujita H, Otsuka T, Aishima S, Nagai E, Oda Y, Tanaka M. MicroRNA-10b is overexpressed in pancreatic cancer, promotes its invasiveness, and correlates with a poor prognosis. Surgery. 2011; 150:916–22.

42. Liu Y, Zhou J, Zhang C, Fu W, Xiao X, Ruan S, Zhang Y, Luo X, Tang M. HLJ1 is a novel biomarker for colorectal carcinoma progression and overall patient survival. Int J Clin Exp Pathol. 2014; 7:969–77.

43. Zhang GJ, Li Y, Zhou H, Xiao HX, Zhou T. miR20a is an independent prognostic factor in colorectal cancer and is involved in cell metastasis. Mol Med Rep. 2014; 10:283–91.

44. Chai H, Liu M, Tian R, Li X, Tang H. miR-20a targets BNIP2 and contributes chemotherapeutic resistance in colorectal adenocarcinoma SW480 and SW620 cell lines. Acta Biochim Biophys Sin (Shanghai). 2011; 43:217–25.

45. Choi S, Korstanje R. Proprotein convertases in high-density lipoprotein metabolism. Biomark Res. 2013; 1:27.

46. Wang JL, Lin YW, Chen HM, Kong X, Xiong H, Shen N, Hong J, Fang JY. Calcium prevents tumorigenesis in a mouse model of colorectal cancer. PLoS ONE. 2011; 6:e22566.

47. Letellier E, Schmitz M, Baig K, Beaume N, Schwartz C, Frasquilho S, Antunes L, Marcon N, Nazarov PV, Vallar L, Even J, Haan S. Identification of SOCS2 and SOCS6 as biomarkers in human colorectal cancer. Br J Cancer. 2014; 111:726–35.

48. Jiang Y, Meng Q, Qi J, Shen H, Sun S. MiR-497 promotes metastasis of colorectal cancer cells through Nrdp1 inhibition. Tumour Biol. 2015; 36:7641–7.

49. Qian XL, Li YQ, Gu F, Liu FF, Li WD, Zhang XM, Fu L. Overexpression of ubiquitous mitochondrial creatine kinase (uMtCK) accelerates tumor growth by inhibiting apoptosis of breast cancer cells and is associated with a poor prognosis in breast cancer patients. Biochem Biophys Res Commun. 2012; 427:60–6.

50. Tsukamoto S, Ishikawa T, Iida S, Ishiguro M, Mogushi K, Mizushima H, Uetake H, Tanaka H, Sugihara K. Clinical significance of osteoprotegerin expression in human colorectal cancer. Clin Cancer Res. 2011; 17:2444–50.

51. Uhlen M, Oksvold P, Fagerberg L, Lundberg E, Jonasson K, Forsberg M, Zwahlen M, Kampf C, Wester K, Hober S, Wernerus H, Björling L, Ponten F. Towards a knowledge-based Human Protein Atlas. Nat Biotechnol. 2010; 28:1248–50.

52. Nanjappa V, Thomas JK, Marimuthu A, Muthusamy B, Radhakrishnan A, Sharma R, Ahmad Khan A, Balakrishnan L, Sahasrabuddhe NA, Kumar S, Jhaveri BN, Sheth KV, Kumar Khatana R, et al. Plasma Proteome Database as a resource for proteomics research: 2014 update. Nucleic Acids Res. 2014; 42:D959–D965.

53. World Health Organization. WHO International Histological Classification of Tumors, Vol 1–25. Geneva, 1967–1981; 2nd edn, Berlin: Springer-Verlag, 1988-. 1992.

54. Greene FL. Current TNM staging of colorectal cancer. Lancet Oncol. 2007; 8:572–3.

55. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, Braisted J, Klapa M, Currier T, Thiagarajan M, Sturn A, Snuffin M, Rezantsev A, et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques. 2003; 34:374–8.

56. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001; 98:5116–21.

57. Dweep H, Sticht C, Pandey P, Gretz N. miRWalk--database: prediction of possible miRNA binding sites by “walking” the genes of three genomes. J Biomed Inform. 2011; 44:839–47.

58. Xiao F, Zuo Z, Cai G, Kang S, Gao X, Li T. miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Res. 2009; 37(Database issue):D105-D110.

59. Risueno A, Fontanillo C, Dinger ME, De las RJ. GATExplorer: genomic and transcriptomic explorer; mapping expression probes to gene loci, transcripts, exons and ncRNAs. BMC Bioinformatics. 2010; 11:221.

60. Gabriel KR, Odoroff CL. Biplots in biomedical research. Stat Med. 1990; 9:469–85.

61. Kim SK, Kim SY, Kim JH, Roh SA, Cho DH, Kim YS, Kim JC. A nineteen gene-based risk score classifier predicts prognosis of colorectal cancer patients. Mol Oncol. 2014; 8:1653–66.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 12140