Skip to main content
Erschienen in: Molecular Cancer 1/2008

Open Access 01.12.2008 | Research

Gene expression profiles in primary pancreatic tumors and metastatic lesions of Ela-c-myc transgenic mice

verfasst von: Archana Thakur, Aliccia Bollig, Jiusheng Wu, Dezhong J Liao

Erschienen in: Molecular Cancer | Ausgabe 1/2008

Abstract

Background

Pancreatic carcinoma usually is a fatal disease with no cure, mainly due to its invasion and metastasis prior to diagnosis. We analyzed the gene expression profiles of paired primary pancreatic tumors and metastatic lesions from Ela-c-myc transgenic mice in order to identify genes that may be involved in the pancreatic cancer progression. Differentially expressed selected genes were verified by semi-quantitative and quantitative RT-PCR. To further evaluate the relevance of some of the selected differentially expressed genes, we investigated their expression pattern in human pancreatic cancer cell lines with high and low metastatic potentials.

Results

Data indicate that genes involved in posttranscriptional regulation were a major functional category of upregulated genes in both primary pancreatic tumors (PT) and liver metastatic lesions (LM) compared to normal pancreas (NP). In particular, differential expression for splicing factors, RNA binding/pre-mRNA processing factors and spliceosome related genes were observed, indicating that RNA processing and editing related events may play critical roles in pancreatic tumor development and progression. High expression of insulin growth factor binding protein-1 (Igfbp1) and Serine proteinase inhibitor A1 (Serpina1), and low levels or absence of Wt1 gene expression were exclusive to liver metastatic lesion samples.

Conclusion

We identified Igfbp1, Serpina1 and Wt1 genes that are likely to be clinically useful biomarkers for prognostic or therapeutic purposes in metastatic pancreatic cancer, particularly in pancreatic cancer where c-Myc is overexpressed.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-7-11) contains supplementary material, which is available to authorized users.
Archana Thakur, Aliccia Bollig contributed equally to this work.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

AT participated in the design of the study; participated in the experimental design; analysis and interpretation of data; and wrote the manuscript; AB designed primers; carried out the semi-quantitative and quantitative RT-PCR; JW isolated RNA from tissue samples and did sequencing; DJL participated in the design of the study, monitored and collected primary or metastatic tumor tissues. All authors read and approved the final manuscript.

Background

Pancreatic cancer (PC) is the fourth leading cause of cancer death in the United States and has no cure, partly because the tumor is at advanced stage or has already metastasized at the time of diagnosis [1]. Like many other types of cancer, pancreatic cancer also shows high frequencies of overexpression and/or amplification of the c-myc oncogene. In one study, 43.5% of primary tumors and 31.6% of metastases showed c-Myc overexpression, in association with 32.5% and 29.4% of gene amplification in the primary and metastatic lesions, respectively [2]. c-Myc and cyclin D1 gene amplification was report 54% and 28% in 31 pancreatic cancer cell lines, respectively, indicating a high frequency of concomitant amplification of both genes [3]. Moreover, simultaneous amplification of activated k-ras and c-myc has been found in both primary tumor and lymph node metastasis, suggesting that c-Myc may collaborate with other oncogenes to promote development and progression of pancreatic cancer [4]. More direct evidence for a critical role for c-Myc in pancreatic carcinogenesis comes from Ela-c-myc transgenic mice that develop PC between 2–7 months of age with 100% incidence rate [5]. One-half of the pancreatic tumors that form in this mouse model are acinar cell adenocarcinomas, while the remaining half of the tumors are mixed ductal and acinar cell carcinomas embedded in dense stroma. We have recently described detailed morphological traits of the pancreatic tumors developed in this transgenic model [6, 7] and, for the first time, observed spontaneous metastasis to the liver in this model. These transgenic mice are among the few animal models of liver metastasis of spontaneous PC. The whole carcinogenic process, from initiation to metastasis, is short (in only a few months time) and is initiated by only one gene.
The most devastating aspect of all types of cancer, particularly pancreatic cancer, is the emergence of metastases in organs distant from the primary tumor, and this remains the primary cause for the poor survival of patients with pancreatic cancer [8]. Therefore, a search for molecular markers that can predict poor prognosis and also serve as novel targets for the development of therapies against this most aggressive disease is warranted. Transgenic animals have been widely used to dissect the role of genes and molecular pathways in cancer [9]. Our transgenic model will help in understanding the molecular mechanisms by which metastases are generated, which is crucial for the prevention and treatment of metastatic disease. In this study we attempted to identify genes that may be responsible for the liver metastasis of pancreatic tumors in Ela-myc transgenic mice.

Results

cDNA Microarray Analysis and Global Gene Expression Profiles

Microarray signal values were calculated from the multiple probes present on each chip for each condition and each condition was repeated at least three times. The relative intensity (fold change) of gene expression levels in the primary tumors (PT) compared to the normal pancreas (NP) is shown in Figure 1A (left panel) and fold change in gene expression in liver metastatic (LM) lesions compared to PT are presented in Figures 1A (right panel).
Cluster analysis was used to display the gene expression data of those, which showed 4-fold higher or 4-fold lower expression levels in PT and LM compared to NP samples. Before clustering, a filtering procedure eliminated genes with uniformly low expression or with low expression variation across the replicates. A large number of genes in PT and LM showed different expression from NP. However, the majority of genes did not show obvious distinction in their expression pattern between the PT and LM (Fig. 1B), except for a small number of genes (boxed area in Fig. 1B expanded in Fig. 1C), suggesting that the LM largely retain the properties of the primary tumors.

Identification of potential tumor promoting genes in c-myc-induced pancreatic tumors

Expressed genes were categorized on the basis of their functional properties, which showed at least 4-fold higher, or 4-fold lower expression levels in primary or metastatic pancreatic tumors compared to normal pancreas. Table 1 shows genes whose expression was upregulated in PT compared to NP (relative fold change) and also shows the relative fold change in LM compared to PT samples. Many upregulated genes such as Birc5, Ccna2, Ccnb1, Ccnb2, Mcm7, Nap1l1, Rad51, Smc4l1, Smc2l1, Rsk4, sfrs1, and sfrs2 (please see Table 1 for their full names) showed 5–20 fold higher expression levels, very few showed exceptionally high fold changes, for example calcium binding protein-S100g showed 109 fold higher expression level in PT than in NP. A large number of upregulated genes in PT belonged to the functional categories known for cell proliferation and cell cycle regulation, chromosomal organization and biogenesis, and RNA processing and modification. In Table 2, we show the genes whose expression was down regulated in PT compared to NP samples (relative fold change) as well as the fold change in LM compared to PT samples. Down regulation of some of the genes in Table 2 including Col4a4, Pcdh17, Muc2, Muc13 (please see Table 2 for their full names) has been shown to modulate cell adhesion and apoptosis.
Table 1
Upregulated genes in primary pancreatic tumors. Relative fold change in primary pancreatic tumors compared to normal pancreas (PT/NP) and in liver metastatic lesions compared to primary pancreatic tumors (LM/PT).
Entrez Gene
Fold change LM*/PT*
Fold change PT/NP*
Gene Symbol
Gene description
Ref.*
Mitochondrial ribosomal subunits
77721
1.0
4.2
Mrps5
Mitochondrial ribosomal protein S5
 
69527
1.0
4.5
Mrps9
Mitochondrial ribosomal protein S9
 
94063
1.0
4.1
Mrpl16
Mitochondrial ribosomal protein L16
 
56284
0.9
5.0
Mrpl19
Mitochondrial ribosomal protein L19
 
66407
0.8
4.1
Mrps15
Mitochondrial ribosomal protein S15
 
64655
1.2
7.6
Mrps22
Mitochondrial ribosomal protein S22
 
64658
1.0
4.1
Mrps25
Mitochondrial ribosomal protein S25
 
Nucleolar and nucleosome assembly proteins
53605
0.9
13.5
Nap1l1
Nucleosome assembly protein 1-like 1
10, 11
110109
0.9
4.3
Nol1
Nucleolar protein 1
 
52530
1.0
10.0
Nola2
Nucleolar protein family A, member 2
 
100608
1.1
9.4
Noc4l
Nucleolar complex associated 4 homolog
 
55989
0.8
6.3
Nol5
Nucleolar protein 5
 
67134
0.9
7.8
Nol5a
Nucleolar protein 5A
 
Small nuclear ribonucleoprotein complex
68981
1.1
8.7
Snrpa1
Small nuclear ribonucleoprotein polypeptide A'
 
20638
0.9
8.3
Snrpb
Small nuclear ribonucleoprotein B
 
20641
1.1
7.1
Snrpd1
Small nuclear ribonucleoprotein D1
 
67332
1.1
7.4
Snrpd3
Small nuclear ribonucleoprotein D3
 
69878
1.1
6.9
Snrpf
Small nuclear ribonucleoprotein polypeptide F
 
666609
1.0
7.6
Snrpg
small nuclear ribonucleoprotein polypeptide G
 
Splicing factor
110809
1.1
5.5
Sfrs1
Splicing factor, arginine/serine-rich 1 (ASF/SF2)
 
20382
1.1
5.1
Sfrs2
Splicing factor, arginine/serine-rich 2 (SC-35)
 
20383
1.1
5.0
Sfrs3
Splicing factor, arginine/serine-rich 3 (SRp20)
 
81898
1.2
5.2
Sf3b1
Splicing factor 3b, subunit 1
15
66125
1.2
8.0
Sf3b5
Splicing factor 3b, subunit 5
15
225027
1.2
4.1
Sfrs7
Splicing factor, arginine/serine-rich 7
 
RNA binding and pre-mRNA processing factors
28000
1.1
4.7
Prpf19
PRP19/PSO4 pre-mRNA processing factor 19 homolog
 
68988
1.1
5.0
Prpf31
PRP31 pre-mRNA processing factor 31 homolog (yeast)
 
56194
1.1
5.8
Prpf40a
PRP40 pre-mRNA processing factor 40 homolog A (yeast)
 
56275
0.9
5.5
Rbm14
RNA binding motif protein 14
 
67071
1.0
16.2
Rps6ka6 (Rsk4)
Ribosomal protein S6 kinase polypeptide 6
 
Spliceosome complex
81898
1.2
5.2
Sf3b1
Splicing factor 3b, subunit 1
15
66125
1.2
8.0
Sf3b5
Splicing factor 3b, subunit 5
15
20382
1.1
4.9
Sfrs2
Splicing factor, arginine/serine-rich 2 (SC-35)
 
68981
1.1
8.7
Snrpa1
Small nuclear ribonucleoprotein polypeptide A'
 
20638
0.9
8.3
Snrpb
Small nuclear ribonucleoprotein B
 
20641
1.1
7.1
Snrpd1
Small nuclear ribonucleoprotein D1
 
69878
1.1
6.9
Snrpf
Small nuclear ribonucleoprotein polypeptide F
 
666609
1.0
7.6
Snrpg
small nuclear ribonucleoprotein polypeptide G
 
Cell proliferation and cell cycle regulation related genes
12428
1.0
16.6
Ccna2
Cyclin A2
 
268697
1.2
11.2
Ccnb1
Cyclin B1
 
12429
1.1
17.9
Ccnb1-rs1
Cyclin B1, related sequence 1
 
12442
0.9
17.8
Ccnb2
Cyclin B2
15, 25
12448
1.3
4.9
Ccne2
Cyclin E2
 
12449
0.9
8.9
Ccnf
Cyclin F
 
17216
0.9
9.0
Mcm2
Minichromosome maintenance deficient 2
14
17215
0.9
8.6
Mcm3
Minichromosome maintenance deficient 3
 
17217
1.2
8.6
Mcm4
Minichromosome maintenance deficient 4
10
17218
1.0
11.8
Mcm5
Minichromosome maintenance deficient 5
 
17219
1.1
20.1
Mcm6
Minichromosome maintenance deficient 6
 
17220
0.9
11.0
Mcm7
Minichromosome maintenance deficient 7
14
70024
1.1
6.3
Mcm10
Minichromosome maintenance deficient 10
 
11799
1.0
11.1
Birc5
Baculoviral IAP repeat-containing 5
 
12211
1.0
4.4
Birc6
Baculoviral IAP repeat-containing 6
 
12189
1.0
5.5
Brca1
Breast cancer 1
 
70099
0.9
17.3
Smc4l1
Structural maintenance of chromosomes 4
 
19361
1.0
15.1
Rad51
RAD51 homolog (S. cerevisiae)
 
Cell adhesion and migration
12774
1.1
6.7
Ccr5
Chemokine (C-C motif) receptor 5
 
56492
1.4
6.6
Cldn18
Claudin 18
25
Cell communication and signal trasduction
75590
0.8
30.3
Dusp9
Dual specificity phosphatase 9
 
67071
1.0
16.2
Rps6ka6 (Rsk4)
Ribosomal protein S6 kinase polypeptide 6
 
12774
1.1
6.7
Ccr5
Chemokine (C-C motif) receptor 5
 
56275
0.9
5.5
Rbm14
RNA binding motif protein 14
 
12309
0.7
109.4
S100g
S100 calcium binding protein G
10, 25
Apoptosis regulation related
11799
1.0
11.1
Birc5
Baculoviral IAP repeat-containing 5
16
17218
1.0
11.8
Mcm5
Minichromosome maintenance deficient 5,
 
17319
1.1
6.8
Mif
Macrophage migration inhibitory factor
 
Chromosome organization and biogenesis
14211
1.1
12.7
Smc2l1
Structural maintenance of chromosomes 2
 
70099
0.9
17.3
Smc4l1
Structural maintenance of chromosomes 4
 
226026
1.0
5.4
Smc5l1
Structural maintenance of chromosomes 5
 
19361
1.0
15.1
Rad51
RAD51 homolog (S. cerevisiae)
12
12189
1.0
5.5
Brca1
Breast cancer 1
 
53605
0.9
13.5
Nap1l1
Nucleosome assembly protein 1-like 1
10, 11
17216
0.9
9.0
Mcm2
Minichromosome maintenance deficient 2 mitotin
 
17218
1.0
11.8
Mcm5
Minichromosome maintenance deficient 5
 
Transcriptional regulator
22431
0.6
2.7
Wt1
Wilms' tumor suppressor gene
57
NP = Normal pancreas; PT = Primary pancreatic tumor; LM = liver metastatic lesion; Ref.* = References identifying genes previously shown to have deregulated expression in pancreatic cancer
Table 2
Downregulated genes in primary pancreatic tumors. Relative fold change in primary pancreatic tumors compared to normal pancreas (PT/NP) and in liver metastatic lesions compared to primary pancreatic tumors (LM/PT)
Entrez Gene#
Fold change LM/PT
Fold change PT/NP
Gene Symbol
Gene description
Ref.*
Cell adhesion, motility and migration
12340
0.84
-11.6
Capza1
Capping protein (actin filament) muscle Z-line, alpha 1
16
12829
0.98
-10.8
Col4a4
Procollagen, type IV, alpha 4
 
13643
1.02
-7.6
Efnb3
Ephrin B3
 
215384
1.03
-8
Fcgbp
Fc fragment of IgG binding protein
 
16855
1.00
-6.4
Lgals4
Lectin, galactose binding, soluble 4
 
17831
1.02
-40
Muc2
Mucin 2
 
219228
1.51
-18.8
Pcdh17
Protocadherin 17
 
68799
1.20
-7.2
Rgmb
RGM domain family, member B
 
16855
1.00
-6.4
Lgals4
Lectin, galactose binding, soluble 4
 
Cell communication and signal trasduction
12154
1.09
-4
Bmp10
Bone morphogenetic protein 10
 
13643
1.02
-7.6
Efnb3
Ephrin B3
 
14463
1.01
-8
Gata4
GATA binding protein 4
 
15874
0.96
-40
Iapp
Islet amyloid polypeptide
 
16333
0.85
-23.2
Ins1
Insulin I
 
14526
0.91
-21.6
Gcg
Glucagon
 
70497
0.86
-8
Arhgap17
Rho GTPase activating protein 17
 
232201
0.83
-7.6
Arhgap25
Rho GTPase activating protein 25
 
110052
1.00
-8.4
Dek
DEK oncogene (DNA binding)
16
14915
0.98
-13.6
Guca2a
Guanylate cyclase activator 2a (guanylin)
 
212307
0.81
-7.2
Mapre2
Microtubule-associated protein, RP/EB family, member 2
 
20844
1.15
-13.6
Stam
Signal transducing adaptor molecule
 
66042
0.85
-14.8
Sostdc1
Sclerostin domain containing 1
 
68799
1.20
-7.2
Rgmb
RGM domain family, member B
 
80718
0.91
-6.4
Rab27b
RAB27b, member RAS oncogene family
 
18386
0.93
-6
Oprd1
Opioid receptor, delta 1
 
67709
0.88
-13.6
Reg4
Regenerating islet-derived family, member 4
 
Cell cycle and cell proliferation
76499
1.02
-8.8
Clasp2
CLIP associating protein 2
 
16333
0.85
-23.2
Ins1
Insulin I
 
16334
0.98
-40
Ins2
Insulin II
 
212307
0.81
-7.2
Mapre2
Microtubule-associated protein, RP/EB family, member 2
 
22268
0.90
-6
Upk1b
Uroplakin 1B
 
14526
0.91
-21.6
Gcg
Glucagon
 
212307
0.81
-7.2
Mapre2
Microtubule-associated protein, RP/EB family, member 2
 
57263
1.11
-28
Retnlb
Resistin like beta
 
12154
1.09
-4
Bmp10
Bone morphogenetic protein 10
 
17831
1.02
-40
Muc2
Mucin 2
19
17063
0.91
-60
Muc13
Mucin 13, epithelial transmembrane
 
Transporter and binding activity
11773
1.09
-14.8
Ap2m1
Adaptor protein complex AP-2, mu1
 
80718
0.91
-6.4
Rab27b
RAB27b, member RAS oncogene family
 
56185
1.00
-19.2
Hao3
Hydroxyacid oxidase (glycolate oxidase) 3
 
110052
1.00
-8.4
Dek
DEK oncogene (DNA binding)
16
12829
0.98
-10.8
Col4a4
Procollagen, type IV, alpha 4
 
16467
1.13
-11.6
Atcay
Ataxia, cerebellar, Cayman type homolog (human)
 
13487
0.95
-20
Slc26a3
Solute carrier family 26, member 3
 
216156
0.92
-4
Wdr18
WD repeat domain 18
 
69008
1.23
-6.4
Cab39l
Calcium binding protein 39-like
 
12351
0.84
-4
Car4
Carbonic anhydrase 4
 
72832
0.93
-14.8
Crtac1
Cartilage acidic protein 1
 
75600
1.20
-8
Calml4
Calmodulin-like 4
 
Apoptosis
15874
0.96
-40
Iapp
Islet amyloid polypeptide
 
17831
1.02
-40
Muc2
Mucin 2
19
71361
1.15
-8
Amid
Apoptosis-inducing factor, mitochondrion-associated 2
 
16334
0.98
-40
Ins2
Insulin II
 
17063
0.91
-60
Muc13
Mucin 13, epithelial transmembrane
 
Transcription activity
109275
0.94
-4
Actr5
ARP5 actin-related protein 5 homolog (yeast)
 
71458
0.89
-6
Bcor
Bcl6 interacting corepressor
 
14463
1.01
 
Gata4
GATA binding protein 4
 
Epigenetic and chromatin modification
213742
1.00
-8.8
Xist
Inactive X specific transcripts
 
75796
0.86
-4
Cdyl2
Chromodomain protein, Y chromosome-like 2
 
Inflammatory and immune response
21786
0.90
-10.8
Tff3
Trefoil factor 3, intestinal
 
15101
0.90
-7.6
H60
Histocompatibility 60
 
94071
1.00
-4
Clec2h
C-type lectin domain family 2, member h
 
Cell differentiation
12154
1.09
-4
Bmp10
Bone morphogenetic protein 10
 
14463
1.01
-8
Gata4
GATA binding protein 4
 
72324
0.86
-4
Plxdc1
Plexin domain containing 1
 
20755
1.31
-16
Sprr2a
Small proline-rich protein 2A
 
22268
0.90
-6
Upk1b
Uroplakin 1B
 
75770
0.85
-8.4
Brsk2
BR serine/threonine kinase 2
 
Maintenance of cell polarity and shape
76499
1.02
-8.8
Clasp2
CLIP associating protein 2
 
20755
1.31
-16
Sprr2a
Small proline-rich protein 2A
 
Ref.* = References identifying genes previously shown to have deregulated expression in pancreatic cancer
Selected genes (highlighted in Table 1, 2 and 3) from various functional categories were further verified by RT-PCR for their expression patterns (Fig. 2A). This selection was based on results in the literature indicating a direct or indirect role for each candidate gene in RNA processing, cell signaling, cell proliferation or apoptosis and cell adhesion and motility activities resulting in tumor growth and tumor progression. Many of these genes listed in Table 1and 2, such as Birc5, Brca1, Ccnb2, CXCR4, Mcm2, Mcm4, Mcm7, Nap1l1, Rad51, Sf3b, S100g [1017]have been shown to be upregulated, while Cldn18, Muc2, Muc13, and b-myc [1821]are shown to be down regulated in human pancreatic cancer as well as other types of cancer (please see Table 1 and 2for their full names). However, strong expression of Muc13 in 50% of samples as well as b-myc in pancreatic cancer cells was unexpected and needs further characterization.
Table 3
Upregulated genes in liver metastatic lesions. Relative fold change in liver metastatic lesions compared to primary pancreatic tumors (LM/PT) and in primary pancreatic tumors compared to normal pancreas (PT/NP)
Entrez Gene #
Fold change LM/PT
Fold change PT/NP
Gene Symbol
Gene description
Ref.*
Transporter activity
27413
5.1
0.6
Abcb11
ATP-binding cassette, sub-family B (MDR/TAP), member 11
 
12870
11.8
0.9
Cp
Ceruloplasmin
 
107141
4.2
1.1
Cyp2c37
Cytochrome P450, family 2. subfamily c, polypeptide 37
 
76279
9.1
0.6
Cyp2d26
Cytochrome P450, family 2. subfamily d, polypeptide 26
 
13107
7.8
0.3
Cyp2f2
Cytochrome P450, family 2, subfamily f, polypeptide 2
 
14263
11.3
0.4
Fmo5
Flavin containing monooxygenase 5
 
268756
9.0
0.5
Gulo
Gulonolactone (L-) oxidase
 
20493
8.3
0.3
Slc10a1
Solute carrier family 10 member 1
 
69354
8.3
1.0
Slc38a4
Solute carrier family 38, member 4
 
28253
4.9
0.9
Slco1b2
Solute carrier organic anion transporter family, member 1b2
 
Cellular metabolism
67758
10.6
0.3
Aadac
Arylacetamide deacetylase (esterase)
 
208665
11.4
0.3
Akr1d1
Aldo-keto reductase family 1, member D1
 
11806
43.3
0.8
Apoa1
Apolipoprotein A-I
 
238055
12.2
0.6
Apob
Apolipoprotein B
 
12116
33.0
0.6
Bhmt
Betaine-homocysteine methyltransferase
 
14121
9.3
0.7
Fbp1
Fructose bisphosphatase 1
 
227231
33.6
0.3
Cps1
Carbamoyl-phosphate synthetase 1
 
231396
14.8
1.0
Ugt2b36
UDP glucuronosyltransferase 2 family, polypeptide B36
 
15233
6.9
0.4
Hgd
Homogentisate 1, 2-dioxygenase
 
15483
4.2
0.2
Hsd11b1
Hydroxysteroid 11-beta dehydrogenase 1
 
13850
7.8
0.4
Ephx2
Epoxide hydrolase 2, cytoplasmic
 
13077
7.0
0.8
Cyp1a2
Cytochrome P450, family 1, subfamily a, polypeptide 2
 
54150
18.2
0.5
Rdh7
Retinol dehydrogenase 7
 
72094
7.4
1.0
Ugt2a3
UDP glucuronosyltransferase 2 family, polypeptide A3
 
103149
6.3
0.6
Upb1
Ureidopropionase, beta
 
16922
5.4
0.4
Phyh
Phytanoyl-CoA hydroxylase
 
Calcium binding activity
19733
11.6
0.5
Rgn
Regucalcin
 
14067
6.9
0.5
F5
Coagulation factor V
 
16426
48.0
1.0
Itih3
Inter-alpha trypsin inhibitor, heavy chain 3
 
Cell organization and biogenesis
11625
40.5
0.9
Ahsg
Alpha-2-HS-glycoprotein
 
19699
5.5
0.5
Reln
Reelin
 
16008
6.0
1.0
Igfbp2
Insulin-like growth factor binding protein 2
 
14080
74.7
1.0
Fabp1
Fatty acid binding protein 1, liver
 
Protease Inhibitor activity
20700
24.9
4.1
Serpina1a
Serine (or cysteine) peptidase inhibitor, clade A, member 1a
25, 51
20702
100.1
0.4
Serpina1c
Serine (or cysteine) peptidase inhibitor, clade A, member 1c
 
59083
22.8
0.3
Fetub
Fetuin beta
 
Inflammatory and Immune response
12628
4.4
1.1
Cfh
Complement component factor h
 
17175
4.5
1.0
Masp2
Mannan-binding lectin serine peptidase 2
 
11625
40.5
0.9
Ahsg
Alpha-2-HS-glycoprotein
 
15439
14.4
7.6
Hp
Haptoglobin
 
18405
15.8
1.4
Orm1
Orosomucoid 1
 
12583
8.4
0.8
Cdo1
Cysteine dioxygenase 1, cytosolic
 
13850
7.8
0.4
Ephx2
Epoxide hydrolase 2, cytoplasmic
 
11699
90.2
0.2
Ambp
Alpha 1 microglobulin/bikunin
28
Cell Adhesion
12558
4.7
1.0
Cdh2
Cadherin 2
 
14067
6.9
0.5
F5
Coagulation factor V
 
16008
6.0
1.0
Igfbp2
Insulin-like growth factor binding protein 2
 
19699
5.5
0.5
Reln
Reelin
 
17175
4.5
1.0
Masp2
Mannan-binding lectin serine peptidase 2
 
14080
74.7
1.0
Fabp1
Fatty acid binding protein 1, liver
 
Cell growth and cell cycle
14080
74.7
1.0
Fabp1
Fatty acid binding protein 1, liver
 
16008
6.0
1.0
Igfbp2
Insulin-like growth factor binding protein 2
 
11625
40.5
0.9
Ahsg
Alpha-2-HS-glycoprotein
 
Cell motility and migration
12558
4.7
1.0
Cdh2
Cadherin 2
 
19699
5.5
0.5
Reln
Reelin
 
16841
4.8
0.6
Lect2
Leukocyte cell-derived chemotaxin 2
 
20315
4.5
0.1
Cxcl12
Chemokine (C-X-C motif) ligand 12
 
12738
2.8
0.3
Cldn2
Claudin 2
 
Cell communication and Signal Transduction
208665
11.4
0.3
Akr1d1
Aldo-keto reductase family 1, member D1
 
22139
38.8
0.1
Ttr
Transthyretin
 
16008
6.0
1.0
Igfbp2
Insulin-like growth factor binding protein 2
 
20526
13.1
0.3
Slc2a2
Solute carrier family 2, member 2
 
238055
12.2
0.6
Apob
Apolipoprotein B
 
50765
4.4
0.7
Trfr2
Transferrin receptor 2
 
107146
4.7
0.7
Glyat
Glycine-N-acyltransferase
 
51811
5.7
0.7
Clec4f
C-type lectin domain family 4, member f
 
14080
74.7
1.0
Fabp1
Fatty acid binding protein 1, liver
 
56720
4.0
0.8
Tdo2
Tryptophan 2,3-dioxygenase
 
11625
40.5
0.9
Ahsg
Alpha-2-HS-glycoprotein
 
353283
4.1
42.0
Eras
ES cell-expressed Ras
 
19699
5.5
0.5
Reln
Reelin
 
16006
28.1
0.7
Igfbp1
Insulin-like growth factor binding protein 1
28,30,31
Ref.* = References identifying genes previously shown to have deregulated expression in pancreatic cancer
We evidenced notable changes in the family members of insulin-like growth factor (Igf). While Igf1 expression was slightly decreased in tumors compared with normal pancreas in the wild type littermates, Igf2 expression was dramatically increased (Fig 3A). All three receptors for Igf1 and Igf2 showed only slight increase in their expression, on the other hand all Igf binding proteins (Igfbp1, Igfbp2, Igfbp3, Igf2bp1 etc.) were downregulated compared to normal pancreas. Western blot analysis confirmed increased expression of cleaved, active form of Igf2 (Fig 3B).

Identification of potential metastasis promoting genes in c-myc induced pancreatic tumors

As mentioned above, we identified a small number of genes that were under various functional categories in metastatic tissues, which were either significantly upregulated or downregulated compared to PT. Interestingly, genes that were downregulated in liver metastatic lesions were comparatively much fewer than upregulated genes. Table 3 shows 4-fold higher and Table 4, 4-fold lower expression levels in LM compared to PT. Most of the highly upregulated genes such as Cp, Apoa1, Ttr in liver metastatic lesions are known biomarkers for the detection of ovarian or other types of cancer [2224]. Other highly upregulated genes were related to protease inhibition such as Serpina1a, Serpina1c, Ambp [2527]and insulin growth factor binding proteins such as Igfbp1 and Ifgbp2 [2831], which have been shown to be upregulated in human pancreatic cancer as well as in the animal models of either pancreatic cancer or other types of cancer. For the verification of some of these genes, we selected two upregulated and two downregulated genes, that showed striking differences from primary pancreatic tumors. In line with our mocroarray data, all LM samples verified by RT-PCR showed highly consistent results (Figure 2B).
Table 4
Downregulated genes in liver metastatic lesions. Relative fold change in liver metastatic lesions compared to primary pancreatic tumors (LM/PT) and in primary pancreatic tumors compared to normal pancreas (PT/NP)
Entrez Gene #
LM/PT
PT/NP
Gene Symbol
Gene description
Ref.*
Cell communication and Signal transduction
22329
0.5
23.5
Vcam1
Vascular cell adhesion molecule 1
 
58194
0.4
4.0
Sh3kbp1
SH3-domain kinase binding protein 1
 
15186
0.1
15.0
Hdc
Histidine decarboxylase
 
11438
0.2
4.9
Chrna4
Cholinergic receptor, nicotinic, alpha polypeptide 4
 
12524
0.6
4.6
Cd86
CD86 antigen
 
93761
0.2
4.2
Smarca1
SWI/SNF related, regulator of chromatin, subfamily a, member 1
 
Cell motility and migration
12767
0.7
4.7
Cxcr4
Chemokine (C-X-C motif) receptor 4
25
17381
2.8
7.6
Mmp12
Matrix metallopeptidase 12
16
11438
0.2
4.9
Chrna4
Cholinergic receptor, nicotinic, alpha polypeptide 4
 
Cell Adhesion
12505
0.6
5.3
Cd44
CD44 antigen
11
22329
0.5
23.5
Vcam1
Vascular cell adhesion molecule 1
 
Cell death and apoptosis
18616
0.2
11.2
Peg3
Paternally expressed 3
 
11801
0.6
31.1
Cd5l
CD5 antigen-like
 
58194
0.4
4.0
Sh3kbp1
SH3-domain kinase binding protein 1
 
Inflammatory and Immune response
20210
0.1
14.1
Saa3
Serum amyloid A 3
 
58194
0.4
4.0
Sh3kbp1
SH3-domain kinase binding protein 1
 
15186
0.1
15.0
Hdc
Histidine decarboxylase
 
Ref.* = References identifying genes previously shown to have deregulated expression in pancreatic cancer

Decreased or lost expression of Wt1 mRNA in primary pancreatic tumors

Wt1 is a transcription factor and has been found to be overexpressed in several types of cancers with poor prognosis. Our microarray data showed two-fold higher expression of the Wt1 gene in PT samples compared to NP samples. RT-PCR with a pair of primers that amplify exons 1 to 7 could detect Wt1 mRNA in PT but not in NP and LM (Fig. 2D). Interestingly, liver metastatic lesions expressed a lower molecular species of mRNA. We purified the higher band from primary tumors and the lower band from liver metastatic lesions and sequenced the PCR products. The results showed that the Wt1 mRNA in PT contained both wild type Wt1 and Wt1 variant without exon 5 (-51 nt). The slight difference in length could be visualized on agarose gel when the PCR products were separated further (Fig. 2D, amplified zone). On the other hand, sequencing results of the band in liver metastatic lesions showed that it was a product of Uroc1 (urocanase domain containing 1) gene, not Wt1. Comparison of the primer sequences with the mouse Uroc1 cDNA (NM_144940) showed high homology, and therefore a non-specific band (Uroc1) was amplified with this primer pair. Since human Uroc1 gene is highly expression in hepatoblastoma than in fetal liver [32], it is possible that Uroc1 is preferentially expressed in liver tumors and thus may serve as a marker. PCR with another pair of primers that amplified nt1444-1943 region of the mRNA also showed that LM expressed much lower levels of Wt1. Considering that a tissue is heterogeneous in cell types, it is reasonable to assume that the Wt1 mRNA detected in LM was derived from stromal tissue whereas the cancer cells might have lost Wt1 expression.

Real-time Quantitative Reverse Transcription-PCR Validation

To confirm the array gene expression data, we performed quantitative reverse transcription-PCR (qRT-PCR) for a selected set (n = 10) of genes and the representative data for three genes are shown in Table 4. Although the extent of measured values detected by the two methods varied, an overall pattern concordance between qRT-PCR and Affymetrix cDNA array experiments was observed (i.e., same trend of induction or suppression was detected by both methods for each target genes). This difference may be due to probe design or the GeneChip system hybridization conditions. For all qRT-PCR, primers specific to β-actin were used as a control to normalize each experiment. Results are presented in Table 5.
Table 5
Quantitative RT-PCR. Relative quantity of mRNA expression in PT, LM and NP tissues measured by quantitative real time PCR
 
Relative fold change
Genes
PT1
LT1
PT2
LT2
PT3
LT3
NP1
NP2
Igfbp1
14.4
70.0
2.0
20.0
10.0
90.0
2.0
2.0
Sepina1a
16.9
4.9
28.9
78.4
14.4
40.0
2.0
2.0
Peg3
0.4
0.2
4.9
10.0
16.0
8.1
2.0
2.0
β-actin
1.0
1.0
1.0
1.0
1.0
1.0
1.0
1.0

Verification of microarray data in human pancreatic cancer cell lines

A panel of human pancreatic cancer cell lines that were reportedly to have high or low metastatic potential in immunodeficient mouse models were used to verify the data from Ela-c-myc model of primary and metastatic pancreatic tumors. Cell lines with high metastatic potential include PANC28, CoLo357fg, L3.6pl and low- or non-metastatic potential include PANC1 and BxPC3. We verified two genes in human cell lines, Igfbp1 and Serpina1a, these genes were highly upregulated in liver metastaic tissues compared to primary pancreatic tumors from transgenic mice. Expression patterns of both genes were consistent with the murine microarray and RT-PCR data (Fig. 2C).

Discussion

In this study, we report the genome-wide expression profiles of primary pancreatic tumors and liver metastatic lesions from Ela-c-myc transgenic mice, or normal pancreas from wild-type mice. cDNA microarray analysis showed several gene clusters under various functional categories in primary or metastatic pancreatic tumors of Ela-c-myc transgenic mice that differ from normal pancreas of non-transgenic littermates. Notably, increased expression was observed for a large number of genes related to ribosomal biogenesis, maturation and ribosome assembly in primary or metastatic pancreatic tumors. Previous studies by others have also shown enhanced expression of genes related to ribosomal proteins, rRNA maturation and ribosome assembly, in addition to enhanced expression of many translation initiation and elongation factors in c-Myc overexpressing cells [3335]. Thus, our model recapitulates the experimental observations and key features of c-Myc overexpressing tumors.
Genes involved in posttranscriptional regulation was a major functional category of upregulated genes in both PT and LM compared to NP samples, we observed changes in expression for splicing factors, RNA binding/pre-mRNA processing factors and spliceosome related genes, indicating that events related to RNA processing may play critical roles in pancreatic tumor development and progression induced by c-Myc. More than 50% of human genes undergo alternative splicing, and this type of RNA process has recently become an emerging topic in molecular and clinical oncology [3638]. Our data showed upregulation of several splicing factors from the SR family such as Sfrs1, Sfrs2, Sfrs3, Sf3b in both primary and metastatic tumors compared to normal pancreas. SR proteins represent a family of essential splicing factors, which are characterized by extensively phosphorylated serine-arginine rich domains [39]. SR proteins recognize splice sites and, depending on their relative levels, these proteins can influence alternative RNA processing [40].
Other groups of genes that were upregulated are involved in DNA replication, cell proliferation and cell cycle regulation; chromosome organization and biogenesis; and signal transduction. Many genes are related to the maintenance of chromosomal structure and integrity such as minichromosome maintenance (Mcm)2, Mcm5, Mcm10, structural maintenance of chromosome (Smc)2l1, Smc4l1, Smc5l1, Rad51, Brca1 and Centromere component (Cenp-I). The entire Mcm protein family (Mcm2-7) is essential in regulating the replication of DNA. Amplification of genes in the Mcm family has been detected in various cancer cells [41]. Their upregulation may deregulate the complete and accurate DNA replication and thus result in failure to maintain the genetic integrity of affected cells. Smc family proteins are integral components of the machinery that modulates chromosome structure for mitosis [42]. Similarly, Rad51, brca1 and Cenp-I play a role in maintenance of genetic integrity [43, 44]. We also noticed increased expression of some X-linked genes related to signal transduction such as Rsk4, Dusp9 and S100g, which have not been reported previously in pancreatic tumors.
Intriguingly, we observed highly upregulated expression of Igfbp1 and Serpina1 in liver metastatic tissues compared to primary pancreatic tumors and normal pancreas. Verification of Igfbp1 and Serpina1 by RT-PCR and quantitative PCR showed strong expression in liver metastatic lesions but there was a lack of expression of these genes in primary pancreatic tumors or normal pancreas. Similarly, both these genes also showed higher expression in highly metastatic human pancreatic cell lines (PANC28, CoLo357fg, L3.6pl) and lower expression levels in less-metastatic cell lines (PANC1 and BxPC3). Several studies have described the inhibitory and potentiating activities of both Serpina1 and Igfbp1 in a variety of cells [4547]. Igfbp1 interacts with α5β1 integrin, influencing cell adhesion and migration. Jones et al. [48] first reported the increased migration of Chinese hamster ovary cells transfected to express human Igfbp1. Increased expression of several Igfbps has also been reported in human pancreatic cancer [2831]. Serpins are endogenous inhibitors of serine protease activity in vivo [49, 50] and a large number of studies support the notion that proteases play an important role in the progression of malignant tumors. Therefore, the expression of proteinase inhibitors is considered to be an anti-malignant event. Serpina1, a major inhibitor of human serine proteases in serum, is produced mainly by the liver, but also by extra-hepatic cells, including neutrophils and certain cancer cells [51, 52]. However, clinical studies have shown that high circulating levels of Serpina1 directly correlate with tumor progression [53, 54]. Immunohistochemical studies revealed that patients with Serpina1-positive lung adenocarcinomas had a worse prognosis than Serpina1-negative ones [55]. More interestingly, both Serpina1 and Igfbp1 have been demonstrated to play a role in human invasive and metastatic pancreatic cancer. Together these studies and our findings suggest that Igfbp1 and Serpina1 may play critical roles in tumor progression in vivo, and are potential candidates for therapeutic interventions.
We also compared our gene expression profiles with published data on human pancreatic cancer tissues or cell lines. Gene expression pattern of many genes such as Serpina1, Igfbp1, Wt1, CD44, MMP12, CXCR4, Muc2, Dek, Capza1, Bcra1, Birc5, S100g, Claudin-18, RAD51, Mcm2, Mcm4, Mcm7, Cyclin B2, splicing factor 3b, Nap1l1 etc. (please see Tables 1, 2, 3 and 4for references) was similarly reported in other studies and therefore provide a validation for our model.

Conclusion

We show differential gene expression profiles under several functional categories in normal pancreas, primary pancreatic tumors and liver metastases. We identified two genes, Igfbp1 and Serpina1, which were overexpressed only in liver metastatic lesions suggesting that these genes are likely to be involved in the establishment of metastases in Ela-myc transgenic animal model. In addition, metastatic lesions appear to have low levels or absence of Wt1 gene expression while primary tumors express at least two major variants (+ exon 5 or - exon 5) Wt1 transcripts. Igfbp1 and Serpina1 may serve as clinically interesting biomarkers are likely to be useful for prognostic or therapeutic purposes in metastatic pancreatic cancer.

Methods

Ela-myc transgenic mice

We used Ela-myc transgenic mice with a FVB background, this strain was generated by crossbreeding of C57BL/6xSJL background Ela-myc [5] mice (obtained from Dr. Sandgren at the University of Wisconsin) with a FVB strain. The F1 mice were crossed together to generate F2 transgenic mice and some of the F2 mice were crossed to yield F3 mice. The F2 and F3 transgenic mice and their wild type littermates were used in this study.

Human Pancreatic cancer cell lines

A panel of human pancreatic cell lines, PANC1, PANC-28, CoLo357, L3.6pl and BxPC3, were used to verify the microarray data. All pancreatic cell lines were cultured in RPMI 1640 supplemented with 10% fetal bovine serum, penicillin and streptomycin. Cells were harvested when they were about 80–90% confluent for RNA isolation.

cDNA microarray

Primary pancreatic cancer tissue, its corresponding liver metastatic lesion and normal pancreatic tissues were used to prepare RNA using the RNeasy mini kit (Qiagen) per manufacturer's instructions. Assurance of quality assessment and microarray analysis were carried out by personnel in the Applied Genomics Technology Center (Center for Molecular Medicine and Genetics, Wayne State University). Briefly, biotin-labeled RNA fragments were produced from 1 μg of RNA by first synthesizing double-stranded cDNA followed by in vitro transcription and fragmentation reactions. A hybridization cocktail, containing the fragmented cRNA, probe array controls, bovine serum albumin, and herring sperm DNA, was prepared and hybridized at 45°C for 16 h to the High Density Mouse Genome M430-2 containing 45101 probesets (Affymetrix Inc., Santa Clara, CA). The hybridized probe array was washed, and bound biotin-labeled cRNA was detected with streptavidin-phycoerythrin conjugate. Each probe array was scanned twice (Hewlett-Packard GeneArray Scanner), the images were overlaid, and the average intensities of each probe cell were compiled. Microarray was repeated three times for each condition (LM, PT, NP).

cDNA microarray data analysis

High density microarray image files were interpreted and quality assessed to Affymetrix standards in GCOS 1.1 as described previously [56]. Expression changes were filtered in DChip for fold change (> 4 fold) between the experiments. Hierarchical clustering was carried out using Dchip and ontological analysis of gene expression was conducted in both OntoExpress in conjunction with curated pathway analysis using the KEGG Biocarta and GeneGo systems. At least three samples from each condition were used for Affymetrix microarray analysis to select candidate genes. Candidate genes were also confirmed with semi-quantitative, quantitative RT-PCR analysis and/or western blot at least 3 times.

Semiquantitative RT-PCR

Total RNA, isolated from the primary or metastatic lesions and normal pancreas of Ela-c-myc transgenic mice, was subjected to first-strand cDNA synthesis using an oligo (dT) primer and Moloney murine leukemia virus (MMLV) reverse transcriptase (Invitrogen). The primer amplified products were separated on ethidium bromide containing 1.2% agarose gels. Primers for the semiquantitative and quantitative detection of target mRNAs are presented in Table 6.
Table 6
List of primer. Primer sets for qRT-PCR and sqRT-PCR
Gene name
Accession No.
Quantitative or sqRT-PCR primer sequence
CXCR4
  
Upstream
D87747
CATGGAACCGATCAGTGTGA (325)*
Downstream
 
TTTCCCAAAGTACCAGTCAGC
MMP2
  
Upstream
NM_008610
CTGTGTTCTTCGCAGGGAAT (433)
Downstream
 
TGTGCAGCGATGAAGATGAT
Snail2
  
Upstream
NM_011415
TTCCTCTGACACTTCATCCAA (474)
Downstream
 
TTGGAGCAGTTTTTGCACTG
E-tcad
  
Upstream
NM_009864
CCTGCCAATCCTGATGAAAT (329)
Downstream
 
TCAGGGA AGGAGCTGAAAGA
Fgf13
  
Upstream
AF020737
CATTTTCTGCCCAAACCACT (378)
Downstream
 
AATGCTTGGCACTCTTTTGC
Rsk4
  
Upstream
BB402211
GTGGGTGCCAAAGTTTTGAT (351)
Downstream
 
CAAACCACATGGAAATCAGG
MIF
  
Upstream
NM_010798.1
ACTACAGTAAGCTGCTGTGTGG (208)
Downstream
 
ATCGCTACCGGTGGATAAAC
Mcm7
  
Upstream
NM_008568.1
ACCGCGAAGTCAGTACACAA (208)
Downstream
 
GATGGTCTGCTGCTCCATAA
Ttr
  
Upstream
NM_013697.1
TGGAAGACACTTGGCATTTC (194)
Downstream
 
TGCTACTGCTTTGGCAAGAT
H2Aa
  
Upstream
NM_010378.2
CCTTCATCCCTTCTGACGAT (197)
Downstream
 
CAGGCCTTGAATGATGAAGA
Mrpl19
  
Upstream
NM_026490.2
TGCATCCCATGAAGAAGAGA (183)
Downstream
 
GACATTTGCTCGTTACAAAAGC
Dusp9
  
Upstream
NM_029352.3
CCTGTGCTTGAGCTCTGATT (181)
Downstream
 
GCTCTCCAAATTGGCTGAAT
S100g
  
Upstream
NM_009789.2
CAGCAAAATGTGTGCTGAGA (197)
Downstream
 
CTCCATCGCCATTCTTATCC
Serpina1a
  
Upstream
NM_009243
GCCCTGGCAAATTACATTCT (196)
Downstream
 
CATTGCCTGCATAATCCATC
Peg3
  
Upstream
NM_008817.2
ACCATTCAGGCCTCAGTTTC (205)
Downstream
 
TTTTCTCAAATTCGCTGACG
Igfbp1
  
Upstream
NM_008341
CCTGCCAACGAGAACTCTAT (196)
Downstream
 
GGGATTTTCTTTCCACTCCA
Saa3
  
Upstream
NM_011315.3
GCGAGCCTACTCTGACATGA (196)
Downstream
 
ATTGGCAAACTGGTCAGCTC
Cldn18
  
Upstream
NM_019815.2
GCTGTACGAGCCCTGATGAT (193)
Downstream
 
TGTTGGCAAACACAGACACA
Sfrs1
  
Upstream
NM_173374.3
CACTGGTGTCGTGGAGTTTG (190)
Downstream
 
CTTCTGCTACGGCTTCTGCT
Sfrs2
  
Upstream
NM_013663.3
GCTTTGCTTTCGTCGAATTT (188)
Downstream
 
AGGACTCCTCCTGCGGTAAT
Eif2
  
Upstream
NM_026030.2
GGAGTTGCTGAACCGAGTGT (180)
Downstream
 
AGGAGATGTTTGGGTTGACG
Muc13
  
Upstream
NM_010739.1
TGCGTGATGCTACAAAGGAC (195)
Downstream
 
TGTCCTGGCATTTACTGCTG
Igfbp1 (human)
  
Upstream
NM_000596.2
AAGGCACAGGAGACATCAGG (195)
Downstream
 
TATCTGGCAGTTGGGGTCTC
Serpina1 (human)
  
Upstream
NM_001002235.1
TGCCTGATGAGGGGAAACTA (186)
Downstream
 
CCCCATTGCTGAAGACCTTA
WT1(362–970)
  
Upstream
NC_000068
TCCAGCAGCCGGAGCAACCT (608)
Downstream
 
AGGGCGTGTGGCCATAGCTG
WT1(947–1470)
  
Upstream
NC_000068
CGCCCAGCTATGGCCACACG (523)
Downstream
 
ATTGCAGCCTGGGTATGCAC
WT1(1444–1943)
  
Upstream
NC_000068
TTCATGTGTGCATACCCAGG (499)
Downstream
 
GTAGATCCACAGTCGTGTCC
*PCR product size

Real-Time RT-PCR

cDNA from the primary or metastatic lesions Ela-c-myc transgenic and normal pancreas of wild type mice were subjected to PCR amplification, a maximum of 2 μl of each cDNA sample was used per 25-μl PCR reactions. The real-time measurements were analyzed in triplicate using an automated Real Time Cycler as described previously [56]. The relative quantity in primary tumor versus normal tissue or primary tumor versus metastatic lesion was normalized to β-actin.

Sequencing of Wilm's tumor suppressor gene (Wt1)

RT-PCR analysis using primers amplified nt947-1470 region of mouse Wt1 mRNA, which covers the first 7 exons, showed that liver metastases (but not primary pancreatic tumors) contained a lower molecular weight mRNA species. To verify the identity of the PCR products of the higher bands in primary tumor and lower band in liver metastatic lesions, we sequenced these bands using forward primer-947 after purifying them from agarose gels using Gel Extraction Kit (QIAEX II) from Qiagen.

Acknowledgements

This work was supported by a grant from Elsa U. Pardee Foundation on pancreatic cancer research.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

AT participated in the design of the study; participated in the experimental design; analysis and interpretation of data; and wrote the manuscript; AB designed primers; carried out the semi-quantitative and quantitative RT-PCR; JW isolated RNA from tissue samples and did sequencing; DJL participated in the design of the study, monitored and collected primary or metastatic tumor tissues. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Yeo TP, Hruban RH, Leach SD, Wilentz RE, Sohn TA, Kern SE, Iacobuzio-Donahue CA, Maitra A, Goggins M, Canto MI: Pancreatic cancer. Curr Probl Cancer. 2002, 26 (4): 176-275.CrossRefPubMed Yeo TP, Hruban RH, Leach SD, Wilentz RE, Sohn TA, Kern SE, Iacobuzio-Donahue CA, Maitra A, Goggins M, Canto MI: Pancreatic cancer. Curr Probl Cancer. 2002, 26 (4): 176-275.CrossRefPubMed
2.
Zurück zum Zitat Schleger C, Verbeke C, Hildenbrand R, Zentgraf H, Bleyl U: c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Mod Pathol. 2002, 15 (4): 462-469.CrossRefPubMed Schleger C, Verbeke C, Hildenbrand R, Zentgraf H, Bleyl U: c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Mod Pathol. 2002, 15 (4): 462-469.CrossRefPubMed
3.
Zurück zum Zitat Mahlamaki EH, Barlund M, Tanner M, Gorunova L, Hoglund M, Karhu R, Kallioniemi A: Frequent amplification of 8q24, 11q, 17q, and 20q-specific genes in pancreatic cancer. Genes Chromosomes Cancer. 2002, 35 (4): 353-358.CrossRefPubMed Mahlamaki EH, Barlund M, Tanner M, Gorunova L, Hoglund M, Karhu R, Kallioniemi A: Frequent amplification of 8q24, 11q, 17q, and 20q-specific genes in pancreatic cancer. Genes Chromosomes Cancer. 2002, 35 (4): 353-358.CrossRefPubMed
4.
Zurück zum Zitat Yamada H, Sakamoto H, Taira M, Nishimura S, Shimosato Y, Terada M, Sugimura T: Amplifications of both c-Ki-ras with a point mutation and c-myc in a primary pancreatic cancer and its metastatic tumors in lymph nodes. Jpn J Cancer Res. 1986, 77 (4): 370-375.PubMed Yamada H, Sakamoto H, Taira M, Nishimura S, Shimosato Y, Terada M, Sugimura T: Amplifications of both c-Ki-ras with a point mutation and c-myc in a primary pancreatic cancer and its metastatic tumors in lymph nodes. Jpn J Cancer Res. 1986, 77 (4): 370-375.PubMed
5.
Zurück zum Zitat Sandgren EP, Quaife CJ, Paulovich AG, Palmiter RD, Brinster RL: Pancreatic tumor pathogenesis reflects the causative genetic lesion. Proc Natl Acad Sci USA. 1991, 88 (1): 93-97.PubMedCentralCrossRefPubMed Sandgren EP, Quaife CJ, Paulovich AG, Palmiter RD, Brinster RL: Pancreatic tumor pathogenesis reflects the causative genetic lesion. Proc Natl Acad Sci USA. 1991, 88 (1): 93-97.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Liao DJ, Wang Y, Wu J, Adsay NV, Grignon D, Khanani F, Sarkar FH: Characterization of pancreatic lesions from MT-tgfalpha, Ela-myc and MT-tgfalpha/Ela-myc single and double transgenic mice. J Carcinog. 2006, 5: 19-PubMedCentralCrossRefPubMed Liao DJ, Wang Y, Wu J, Adsay NV, Grignon D, Khanani F, Sarkar FH: Characterization of pancreatic lesions from MT-tgfalpha, Ela-myc and MT-tgfalpha/Ela-myc single and double transgenic mice. J Carcinog. 2006, 5: 19-PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Liao JD, Adsay NV, Khannani F, Grignon D, Thakur A, Sarkar FH: Histological complexities of pancreatic lesions from transgenic mouse models are consistent with biological and morphological heterogeneity of human pancreatic cancer. Histol Histopathol. 2007, 22 (6): 661-676.PubMedCentralPubMed Liao JD, Adsay NV, Khannani F, Grignon D, Thakur A, Sarkar FH: Histological complexities of pancreatic lesions from transgenic mouse models are consistent with biological and morphological heterogeneity of human pancreatic cancer. Histol Histopathol. 2007, 22 (6): 661-676.PubMedCentralPubMed
9.
Zurück zum Zitat Kleeff J, Friess H, Berberat PO, Martignoni ME, Z'Graggen K, Buchler MW: Pancreatic cancer – new aspects of molecular biology research. Swiss Surg. 2000, 6 (5): 231-234.CrossRefPubMed Kleeff J, Friess H, Berberat PO, Martignoni ME, Z'Graggen K, Buchler MW: Pancreatic cancer – new aspects of molecular biology research. Swiss Surg. 2000, 6 (5): 231-234.CrossRefPubMed
10.
Zurück zum Zitat Iacobuzio-Donahue CA, Maitra A, Olsen M, Lowe AW, van Heek NT, Rosty C, Walter K, Sato N, Parker A, Ashfaq R: Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 2003, 162 (4): 1151-1162.PubMedCentralCrossRefPubMed Iacobuzio-Donahue CA, Maitra A, Olsen M, Lowe AW, van Heek NT, Rosty C, Walter K, Sato N, Parker A, Ashfaq R: Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 2003, 162 (4): 1151-1162.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Iacobuzio-Donahue CA, Maitra A, Shen-Ong GL, van Heek T, Ashfaq R, Meyer R, Walter K, Berg K, Hollingsworth MA, Cameron JL: Discovery of novel tumor markers of pancreatic cancer using global gene expression technology. Am J Pathol. 2002, 160 (4): 1239-1249.PubMedCentralCrossRefPubMed Iacobuzio-Donahue CA, Maitra A, Shen-Ong GL, van Heek T, Ashfaq R, Meyer R, Walter K, Berg K, Hollingsworth MA, Cameron JL: Discovery of novel tumor markers of pancreatic cancer using global gene expression technology. Am J Pathol. 2002, 160 (4): 1239-1249.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Maacke H, Jost K, Opitz S, Miska S, Yuan Y, Hasselbach L, Luttges J, Kalthoff H, Sturzbecher HW: DNA repair and recombination factor Rad51 is over-expressed in human pancreatic adenocarcinoma. Oncogene. 2000, 19 (23): 2791-2795.CrossRefPubMed Maacke H, Jost K, Opitz S, Miska S, Yuan Y, Hasselbach L, Luttges J, Kalthoff H, Sturzbecher HW: DNA repair and recombination factor Rad51 is over-expressed in human pancreatic adenocarcinoma. Oncogene. 2000, 19 (23): 2791-2795.CrossRefPubMed
13.
Zurück zum Zitat Lynch HT, Deters CA, Snyder CL, Lynch JF, Villeneuve P, Silberstein J, Martin H, Narod SA, Brand RE: BRCA1 and pancreatic cancer: pedigree findings and their causal relationships. Cancer Genet Cytogenet. 2005, 158 (2): 119-125.CrossRefPubMed Lynch HT, Deters CA, Snyder CL, Lynch JF, Villeneuve P, Silberstein J, Martin H, Narod SA, Brand RE: BRCA1 and pancreatic cancer: pedigree findings and their causal relationships. Cancer Genet Cytogenet. 2005, 158 (2): 119-125.CrossRefPubMed
14.
Zurück zum Zitat Mahlamaki EH, Kauraniemi P, Monni O, Wolf M, Hautaniemi S, Kallioniemi A: High-resolution genomic and expression profiling reveals 105 putative amplification target genes in pancreatic cancer. Neoplasia. 2004, 6 (5): 432-439.PubMedCentralCrossRefPubMed Mahlamaki EH, Kauraniemi P, Monni O, Wolf M, Hautaniemi S, Kallioniemi A: High-resolution genomic and expression profiling reveals 105 putative amplification target genes in pancreatic cancer. Neoplasia. 2004, 6 (5): 432-439.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Nakamura T, Fidler IJ, Coombes KR: Gene expression profile of metastatic human pancreatic cancer cells depends on the organ microenvironment. Cancer Res. 2007, 67 (1): 139-148.CrossRefPubMed Nakamura T, Fidler IJ, Coombes KR: Gene expression profile of metastatic human pancreatic cancer cells depends on the organ microenvironment. Cancer Res. 2007, 67 (1): 139-148.CrossRefPubMed
16.
Zurück zum Zitat Nakamura T, Furukawa Y, Nakagawa H, Tsunoda T, Ohigashi H, Murata K, Ishikawa O, Ohgaki K, Kashimura N, Miyamoto M: Genome-wide cDNA microarray analysis of gene expression profiles in pancreatic cancers using populations of tumor cells and normal ductal epithelial cells selected for purity by laser microdissection. Oncogene. 2004, 23 (13): 2385-2400.CrossRefPubMed Nakamura T, Furukawa Y, Nakagawa H, Tsunoda T, Ohigashi H, Murata K, Ishikawa O, Ohgaki K, Kashimura N, Miyamoto M: Genome-wide cDNA microarray analysis of gene expression profiles in pancreatic cancers using populations of tumor cells and normal ductal epithelial cells selected for purity by laser microdissection. Oncogene. 2004, 23 (13): 2385-2400.CrossRefPubMed
17.
Zurück zum Zitat Soling A, Sackewitz M, Volkmar M, Schaarschmidt D, Jacob R, Holzhausen HJ, Rainov NG: Minichromosome maintenance protein 3 elicits a cancer-restricted immune response in patients with brain malignancies and is a strong independent predictor of survival in patients with anaplastic astrocytoma. Clin Cancer Res. 2005, 11 (1): 249-258.PubMed Soling A, Sackewitz M, Volkmar M, Schaarschmidt D, Jacob R, Holzhausen HJ, Rainov NG: Minichromosome maintenance protein 3 elicits a cancer-restricted immune response in patients with brain malignancies and is a strong independent predictor of survival in patients with anaplastic astrocytoma. Clin Cancer Res. 2005, 11 (1): 249-258.PubMed
18.
Zurück zum Zitat Sanada Y, Oue N, Mitani Y, Yoshida K, Nakayama H, Yasui W: Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J Pathol. 2006, 208 (5): 633-642.CrossRefPubMed Sanada Y, Oue N, Mitani Y, Yoshida K, Nakayama H, Yasui W: Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J Pathol. 2006, 208 (5): 633-642.CrossRefPubMed
19.
Zurück zum Zitat Yonezawa S, Byrd JC, Dahiya R, Ho JJ, Gum JR, Griffiths B, Swallow DM, Kim YS: Differential mucin gene expression in human pancreatic and colon cancer cells. Biochem J. 1991, 276 (Pt 3): 599-605.PubMedCentralCrossRefPubMed Yonezawa S, Byrd JC, Dahiya R, Ho JJ, Gum JR, Griffiths B, Swallow DM, Kim YS: Differential mucin gene expression in human pancreatic and colon cancer cells. Biochem J. 1991, 276 (Pt 3): 599-605.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Hollingsworth MA, Swanson BJ: Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer. 2004, 4 (1): 45-60.CrossRefPubMed Hollingsworth MA, Swanson BJ: Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer. 2004, 4 (1): 45-60.CrossRefPubMed
21.
Zurück zum Zitat Resar LM, Dolde C, Barrett JF, Dang CV: B-myc inhibits neoplastic transformation and transcriptional activation by c-myc. Mol Cell Biol. 1993, 13 (2): 1130-1136.PubMedCentralPubMed Resar LM, Dolde C, Barrett JF, Dang CV: B-myc inhibits neoplastic transformation and transcriptional activation by c-myc. Mol Cell Biol. 1993, 13 (2): 1130-1136.PubMedCentralPubMed
22.
Zurück zum Zitat Griffith OL, Melck A, Jones SJ, Wiseman SM: Meta-analysis and meta-review of thyroid cancer gene expression profiling studies identifies important diagnostic biomarkers. J Clin Oncol. 2006, 24 (31): 5043-5051.CrossRefPubMed Griffith OL, Melck A, Jones SJ, Wiseman SM: Meta-analysis and meta-review of thyroid cancer gene expression profiling studies identifies important diagnostic biomarkers. J Clin Oncol. 2006, 24 (31): 5043-5051.CrossRefPubMed
23.
Zurück zum Zitat Moore LE, Fung ET, McGuire M, Rabkin CC, Molinaro A, Wang Z, Zhang F, Wang J, Yip C, Meng XY: Evaluation of apolipoprotein A1 and posttranslationally modified forms of transthyretin as biomarkers for ovarian cancer detection in an independent study population. Cancer Epidemiol Biomarkers Prev. 2006, 15 (9): 1641-1646.CrossRefPubMed Moore LE, Fung ET, McGuire M, Rabkin CC, Molinaro A, Wang Z, Zhang F, Wang J, Yip C, Meng XY: Evaluation of apolipoprotein A1 and posttranslationally modified forms of transthyretin as biomarkers for ovarian cancer detection in an independent study population. Cancer Epidemiol Biomarkers Prev. 2006, 15 (9): 1641-1646.CrossRefPubMed
24.
Zurück zum Zitat Weinstein PS, Skinner M, Sipe JD, Lokich JJ, Zamcheck N, Cohen AS: Acute-phase proteins or tumour markers: the role of SAA, SAP, CRP and CEA as indicators of metastasis in a broad spectrum of neoplastic diseases. Scand J Immunol. 1984, 19 (3): 193-198.CrossRefPubMed Weinstein PS, Skinner M, Sipe JD, Lokich JJ, Zamcheck N, Cohen AS: Acute-phase proteins or tumour markers: the role of SAA, SAP, CRP and CEA as indicators of metastasis in a broad spectrum of neoplastic diseases. Scand J Immunol. 1984, 19 (3): 193-198.CrossRefPubMed
25.
Zurück zum Zitat Sato N, Fukushima N, Maitra A, Iacobuzio-Donahue CA, van Heek NT, Cameron JL, Yeo CJ, Hruban RH, Goggins M: Gene expression profiling identifies genes associated with invasive intraductal papillary mucinous neoplasms of the pancreas. Am J Pathol. 2004, 164 (3): 903-914.PubMedCentralCrossRefPubMed Sato N, Fukushima N, Maitra A, Iacobuzio-Donahue CA, van Heek NT, Cameron JL, Yeo CJ, Hruban RH, Goggins M: Gene expression profiling identifies genes associated with invasive intraductal papillary mucinous neoplasms of the pancreas. Am J Pathol. 2004, 164 (3): 903-914.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Lian Z, De Luca P, Di Cristofano A: Gene expression analysis reveals a signature of estrogen receptor activation upon loss of Pten in a mouse model of endometrial cancer. J Cell Physiol. 2006, 208 (2): 255-266.CrossRefPubMed Lian Z, De Luca P, Di Cristofano A: Gene expression analysis reveals a signature of estrogen receptor activation upon loss of Pten in a mouse model of endometrial cancer. J Cell Physiol. 2006, 208 (2): 255-266.CrossRefPubMed
27.
Zurück zum Zitat Liu AY, Zhang H, Sorensen CM, Diamond DL: Analysis of prostate cancer by proteomics using tissue specimens. J Urol. 2005, 173 (1): 73-78.CrossRefPubMed Liu AY, Zhang H, Sorensen CM, Diamond DL: Analysis of prostate cancer by proteomics using tissue specimens. J Urol. 2005, 173 (1): 73-78.CrossRefPubMed
28.
Zurück zum Zitat Mauri P, Scarpa A, Nascimbeni AC, Benazzi L, Parmagnani E, Mafficini A, Della Peruta M, Bassi C, Miyazaki K, Sorio C: Identification of proteins released by pancreatic cancer cells by multidimensional protein identification technology: a strategy for identification of novel cancer markers. Faseb J. 2005, 19 (9): 1125-1127.PubMed Mauri P, Scarpa A, Nascimbeni AC, Benazzi L, Parmagnani E, Mafficini A, Della Peruta M, Bassi C, Miyazaki K, Sorio C: Identification of proteins released by pancreatic cancer cells by multidimensional protein identification technology: a strategy for identification of novel cancer markers. Faseb J. 2005, 19 (9): 1125-1127.PubMed
29.
Zurück zum Zitat Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo CJ, Maitra A: Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clin Cancer Res. 2004, 10 (18 Pt 1): 6152-6158.CrossRefPubMed Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo CJ, Maitra A: Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clin Cancer Res. 2004, 10 (18 Pt 1): 6152-6158.CrossRefPubMed
30.
Zurück zum Zitat Karna E, Surazynski A, Orlowski K, Laszkiewicz J, Puchalski Z, Nawrat P, Palka J: Serum and tissue level of insulin-like growth factor-I (IGF-I) and IGF-I binding proteins as an index of pancreatitis and pancreatic cancer. Int J Exp Pathol. 2002, 83 (5): 239-245.PubMedCentralCrossRefPubMed Karna E, Surazynski A, Orlowski K, Laszkiewicz J, Puchalski Z, Nawrat P, Palka J: Serum and tissue level of insulin-like growth factor-I (IGF-I) and IGF-I binding proteins as an index of pancreatitis and pancreatic cancer. Int J Exp Pathol. 2002, 83 (5): 239-245.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Yamada S, Ohira M, Horie H, Ando K, Takayasu H, Suzuki Y, Sugano S, Hirata T, Goto T, Matsunaga T: Expression profiling and differential screening between hepatoblastomas and the corresponding normal livers: identification of high expression of the PLK1 oncogene as a poor-prognostic indicator of hepatoblastomas. Oncogene. 2004, 23 (35): 5901-5911.CrossRefPubMed Yamada S, Ohira M, Horie H, Ando K, Takayasu H, Suzuki Y, Sugano S, Hirata T, Goto T, Matsunaga T: Expression profiling and differential screening between hepatoblastomas and the corresponding normal livers: identification of high expression of the PLK1 oncogene as a poor-prognostic indicator of hepatoblastomas. Oncogene. 2004, 23 (35): 5901-5911.CrossRefPubMed
33.
Zurück zum Zitat Johnson JM, Castle J, Garrett-Engele P, Kan Z, Loerch PM, Armour CD, Santos R, Schadt EE, Stoughton R, Shoemaker DD: Genome-wide survey of human alternative pre-mRNA splicing with exon junction microarrays. Science. 2003, 302 (5653): 2141-2144.CrossRefPubMed Johnson JM, Castle J, Garrett-Engele P, Kan Z, Loerch PM, Armour CD, Santos R, Schadt EE, Stoughton R, Shoemaker DD: Genome-wide survey of human alternative pre-mRNA splicing with exon junction microarrays. Science. 2003, 302 (5653): 2141-2144.CrossRefPubMed
34.
Zurück zum Zitat Pajares MJ, Ezponda T, Catena R, Calvo A, Pio R, Montuenga LM: Alternative splicing: an emerging topic in molecular and clinical oncology. Lancet Oncol. 2007, 8 (4): 349-357.CrossRefPubMed Pajares MJ, Ezponda T, Catena R, Calvo A, Pio R, Montuenga LM: Alternative splicing: an emerging topic in molecular and clinical oncology. Lancet Oncol. 2007, 8 (4): 349-357.CrossRefPubMed
35.
Zurück zum Zitat Srebrow A, Kornblihtt AR: The connection between splicing and cancer. J Cell Sci. 2006, 119 (Pt 13): 2635-2641.CrossRefPubMed Srebrow A, Kornblihtt AR: The connection between splicing and cancer. J Cell Sci. 2006, 119 (Pt 13): 2635-2641.CrossRefPubMed
36.
Zurück zum Zitat Venables JP: Aberrant and alternative splicing in cancer. Cancer Res. 2004, 64 (21): 7647-7654.CrossRefPubMed Venables JP: Aberrant and alternative splicing in cancer. Cancer Res. 2004, 64 (21): 7647-7654.CrossRefPubMed
37.
38.
Zurück zum Zitat Hayes GM, Carrigan PE, Beck AM, Miller LJ: Targeting the RNA splicing machinery as a novel treatment strategy for pancreatic carcinoma. Cancer Res. 2006, 66 (7): 3819-3827.CrossRefPubMed Hayes GM, Carrigan PE, Beck AM, Miller LJ: Targeting the RNA splicing machinery as a novel treatment strategy for pancreatic carcinoma. Cancer Res. 2006, 66 (7): 3819-3827.CrossRefPubMed
39.
Zurück zum Zitat Zahler AM, Neugebauer KM, Lane WS, Roth MB: Distinct functions of SR proteins in alternative pre-mRNA splicing. Science. 1993, 260 (5105): 219-222.CrossRefPubMed Zahler AM, Neugebauer KM, Lane WS, Roth MB: Distinct functions of SR proteins in alternative pre-mRNA splicing. Science. 1993, 260 (5105): 219-222.CrossRefPubMed
40.
Zurück zum Zitat Stickeler E, Kittrell F, Medina D, Berget SM: Stage-specific changes in SR splicing factors and alternative splicing in mammary tumorigenesis. Oncogene. 1999, 18 (24): 3574-3582.CrossRefPubMed Stickeler E, Kittrell F, Medina D, Berget SM: Stage-specific changes in SR splicing factors and alternative splicing in mammary tumorigenesis. Oncogene. 1999, 18 (24): 3574-3582.CrossRefPubMed
41.
Zurück zum Zitat Bailis JM, Forsburg SL: MCM proteins: DNA damage, mutagenesis and repair. Curr Opin Genet Dev. 2004, 14 (1): 17-21.CrossRefPubMed Bailis JM, Forsburg SL: MCM proteins: DNA damage, mutagenesis and repair. Curr Opin Genet Dev. 2004, 14 (1): 17-21.CrossRefPubMed
42.
Zurück zum Zitat Hirano T: At the heart of the chromosome: SMC proteins in action. Nat Rev Mol Cell Biol. 2006, 7 (5): 311-322.CrossRefPubMed Hirano T: At the heart of the chromosome: SMC proteins in action. Nat Rev Mol Cell Biol. 2006, 7 (5): 311-322.CrossRefPubMed
43.
Zurück zum Zitat Amor DJ, Kalitsis P, Sumer H, Choo KH: Building the centromere: from foundation proteins to 3D organization. Trends Cell Biol. 2004, 14 (7): 359-368.CrossRefPubMed Amor DJ, Kalitsis P, Sumer H, Choo KH: Building the centromere: from foundation proteins to 3D organization. Trends Cell Biol. 2004, 14 (7): 359-368.CrossRefPubMed
44.
Zurück zum Zitat Khanna KK, Jackson SP: DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet. 2001, 27 (3): 247-254.CrossRefPubMed Khanna KK, Jackson SP: DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet. 2001, 27 (3): 247-254.CrossRefPubMed
45.
Zurück zum Zitat Kataoka H, Itoh H, Koono M: Emerging multifunctional aspects of cellular serine proteinase inhibitors in tumor progression and tissue regeneration. Pathol Int. 2002, 52 (2): 89-102.CrossRefPubMed Kataoka H, Itoh H, Koono M: Emerging multifunctional aspects of cellular serine proteinase inhibitors in tumor progression and tissue regeneration. Pathol Int. 2002, 52 (2): 89-102.CrossRefPubMed
46.
Zurück zum Zitat Firth SM, Baxter RC: Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev. 2002, 23 (6): 824-854.CrossRefPubMed Firth SM, Baxter RC: Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev. 2002, 23 (6): 824-854.CrossRefPubMed
47.
Zurück zum Zitat Perks CM, Newcomb PV, Norman MR, Holly JM: Effect of insulin-like growth factor binding protein-1 on integrin signalling and the induction of apoptosis in human breast cancer cells. J Mol Endocrinol. 1999, 22 (2): 141-150.CrossRefPubMed Perks CM, Newcomb PV, Norman MR, Holly JM: Effect of insulin-like growth factor binding protein-1 on integrin signalling and the induction of apoptosis in human breast cancer cells. J Mol Endocrinol. 1999, 22 (2): 141-150.CrossRefPubMed
48.
Zurück zum Zitat Jones JI, Doerr ME, Clemmons DR: Cell migration: interactions among integrins, IGFs and IGFBPs. Prog Growth Factor Res. 1995, 6 (2–4): 319-327.CrossRefPubMed Jones JI, Doerr ME, Clemmons DR: Cell migration: interactions among integrins, IGFs and IGFBPs. Prog Growth Factor Res. 1995, 6 (2–4): 319-327.CrossRefPubMed
49.
Zurück zum Zitat Petrache I, Fijalkowska I, Zhen L, Medler TR, Brown E, Cruz P, Choe KH, Taraseviciene-Stewart L, Scerbavicius R, Shapiro L: A novel antiapoptotic role for alpha1-antitrypsin in the prevention of pulmonary emphysema. Am J Respir Crit Care Med. 2006, 173 (11): 1222-1228.PubMedCentralCrossRefPubMed Petrache I, Fijalkowska I, Zhen L, Medler TR, Brown E, Cruz P, Choe KH, Taraseviciene-Stewart L, Scerbavicius R, Shapiro L: A novel antiapoptotic role for alpha1-antitrypsin in the prevention of pulmonary emphysema. Am J Respir Crit Care Med. 2006, 173 (11): 1222-1228.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Zelvyte I, Wallmark A, Piitulainen E, Westin U, Janciauskiene S: Increased plasma levels of serine proteinase inhibitors in lung cancer patients. Anticancer Res. 2004, 24 (1): 241-247.PubMed Zelvyte I, Wallmark A, Piitulainen E, Westin U, Janciauskiene S: Increased plasma levels of serine proteinase inhibitors in lung cancer patients. Anticancer Res. 2004, 24 (1): 241-247.PubMed
51.
Zurück zum Zitat Trichopoulos D, Tzonou A, Kalapothaki V, Sparos L, Kremastinou T, Skoutari M: Alpha 1-antitrypsin and survival in pancreatic cancer. Int J Cancer. 1990, 45 (4): 685-686.CrossRefPubMed Trichopoulos D, Tzonou A, Kalapothaki V, Sparos L, Kremastinou T, Skoutari M: Alpha 1-antitrypsin and survival in pancreatic cancer. Int J Cancer. 1990, 45 (4): 685-686.CrossRefPubMed
52.
Zurück zum Zitat Tzonou A, Sparos L, Kalapothaki V, Zavitsanos X, Rebelakos A, Trichopoulos D: Alpha 1-antitrypsin and survival in hepatocellular carcinoma. Br J Cancer. 1990, 61 (1): 72-73.PubMedCentralCrossRefPubMed Tzonou A, Sparos L, Kalapothaki V, Zavitsanos X, Rebelakos A, Trichopoulos D: Alpha 1-antitrypsin and survival in hepatocellular carcinoma. Br J Cancer. 1990, 61 (1): 72-73.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Higashiyama M, Doi O, Kodama K, Yokouchi H, Tateishi R: An evaluation of the prognostic significance of alpha-1-antitrypsin expression in adenocarcinomas of the lung: an immunohistochemical analysis. Br J Cancer. 1992, 65 (2): 300-302.PubMedCentralCrossRefPubMed Higashiyama M, Doi O, Kodama K, Yokouchi H, Tateishi R: An evaluation of the prognostic significance of alpha-1-antitrypsin expression in adenocarcinomas of the lung: an immunohistochemical analysis. Br J Cancer. 1992, 65 (2): 300-302.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Sun Z, Yang P: Role of imbalance between neutrophil elastase and alpha 1-antitrypsin in cancer development and progression. Lancet Oncol. 2004, 5 (3): 182-190.CrossRefPubMed Sun Z, Yang P: Role of imbalance between neutrophil elastase and alpha 1-antitrypsin in cancer development and progression. Lancet Oncol. 2004, 5 (3): 182-190.CrossRefPubMed
55.
Zurück zum Zitat Higashiyama M, Doi O, Kodama K, Yokouchi H, Tateishi R, Matsuura N, Murata A, Tomita N, Monden T, Ogawa M: Immunohistochemical analysis of pancreatic secretory trypsin inhibitor expression in pulmonary adenocarcinoma: its possible participation in scar formation of the tumor tissues. Tumour Biol. 1992, 13 (5–6): 299-307.CrossRefPubMed Higashiyama M, Doi O, Kodama K, Yokouchi H, Tateishi R, Matsuura N, Murata A, Tomita N, Monden T, Ogawa M: Immunohistochemical analysis of pancreatic secretory trypsin inhibitor expression in pulmonary adenocarcinoma: its possible participation in scar formation of the tumor tissues. Tumour Biol. 1992, 13 (5–6): 299-307.CrossRefPubMed
56.
Zurück zum Zitat Thakur A, Xu H, Wang Y, Bollig A, Biliran H, Liao JD: The role of X-linked genes in breast cancer. Breast Cancer Res Treat. 2005, 93 (2): 135-143.CrossRefPubMed Thakur A, Xu H, Wang Y, Bollig A, Biliran H, Liao JD: The role of X-linked genes in breast cancer. Breast Cancer Res Treat. 2005, 93 (2): 135-143.CrossRefPubMed
57.
Zurück zum Zitat Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, Tatsumi N, Abeno S, Ikeba A, Takashima S, Tsujie M: Overexpression of the Wilms' tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci. 2004, 95 (7): 583-7.CrossRefPubMed Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, Tatsumi N, Abeno S, Ikeba A, Takashima S, Tsujie M: Overexpression of the Wilms' tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci. 2004, 95 (7): 583-7.CrossRefPubMed
Metadaten
Titel
Gene expression profiles in primary pancreatic tumors and metastatic lesions of Ela-c-myc transgenic mice
verfasst von
Archana Thakur
Aliccia Bollig
Jiusheng Wu
Dezhong J Liao
Publikationsdatum
01.12.2008
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2008
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-7-11

Weitere Artikel der Ausgabe 1/2008

Molecular Cancer 1/2008 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.